Elevated plasma triglyceride levels are increasingly considered as an independent risk factor for cardiovascular diseases (
1- Hokanson J.E.
- Austin M.A.
Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies.
,
2- Onat A.
- Sari I.
- Yazici M.
- Can G.
- Hergenç G.
- Avci G.Ş.
Plasma triglycerides, an independent predictor of cardiovascular disease in men: A prospective study based on a population with prevalent metabolic syndrome.
,
3- Sandesara P.B.
- Virani S.S.
- Fazio S.
- Shapiro M.D.
The forgotten lipids: triglycerides, remnant cholesterol, and atherosclerotic cardiovascular disease risk.
). Triglycerides circulate in the blood in two major forms: as chylomicrons carrying the dietary triglycerides and as very low density lipoproteins carrying endogenously produced triglycerides (
4- Goldberg I.J.
- Eckel R.H.
- McPherson R.
Triglycerides and heart disease: still a hypothesis?.
). The clearance of plasma triglycerides is primarily mediated by the action of LPL. This secretory enzyme is produced by parenchymal cells of fat tissue, skeletal muscle, and heart, as well as by macrophages. With the help of the endothelial protein, glycosylphosphatidylinositol-anchored HDL binding protein 1 (GPIHBP1), LPL is transferred from the surface of the sub-endothelial myocytes and adipocytes to the luminal side of the capillary endothelium. There, LPL hydrolyzes the triglycerides contained in the triglyceride-rich lipoproteins to release fatty acids for uptake by the underlying tissues (
5Physiological regulation of lipoprotein lipase.
,
6- Davies B.S.J.
- Beigneux A.P.
- Barnes R.H.
- Tu Y.
- Gin P.
- Weinstein M.M.
- Nobumori C.
- Nyrén R.
- Goldberg I.
- Olivecrona G.
- et al.
GPIHBP1 is responsible for the entry of lipoprotein lipase into capillaries.
,
7- Beigneux A.P.
- Miyashita K.
- Ploug M.
- Blom D.J.
- Ai M.
- Linton M.F.
- Khovidhunkit W.
- Dufour R.
- Garg A.
- McMahon M.A.
- et al.
Autoantibodies against GPIHBP1 as a cause of hypertriglyceridemia.
,
8- Goulbourne C.N.
- Gin P.
- Tatar A.
- Nobumori C.
- Hoenger A.
- Jiang H.
- Grovenor C.R.M.
- Adeyo O.
- Esko J.D.
- Goldberg I.J.
- et al.
The GPIHBP1-LPL complex is responsible for the margination of triglyceride-rich lipoproteins in capillaries.
). The activity of LPL is regulated posttranslationally by numerous factors, many of which are produced in the liver, including several apolipoproteins. In addition, LPL activity is governed by several members of the family of angiopoietin-like proteins (ANGPTLs): ANGPTL3 (
9- Conklin D.
- Gilbertson D.
- Taft D.W.
- Maurer M.F.
- Whitmore T.E.
- Smith D.L.
- Walker K.M.
- Chen L.H.
- Wattler S.
- Nehls M.
- et al.
Identification of a mammalian angiopoietin-related protein expressed specifically in liver.
), ANGPTL4 (
10- Kim I.
- Kim H.G.
- Kim H.
- Kim H.H.
- Park S.K.
- Uhm C.S.
- Lee Z.H.
- Koh G.Y.
Hepatic expression, synthesis and secretion of a novel fibrinogen/angiopoietin-related protein that prevents endothelial-cell apoptosis.
,
11- Kersten S.
- Mandard S.
- Tan N.S.
- Escher P.
- Metzger D.
- Chambon P.
- Gonzalez F.J.
- Desvergne B.
- Wahli W.
Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene.
,
12- Yoon J.C.
- Chickering T.W.
- Rosen E.D.
- Dussault B.
- Qin Y.
- Soukas A.
- Friedman J.M.
- Holmes W.E.
- Spiegelman B.M.
Peroxisome proliferator-activated receptor gamma target gene encoding a novel angiopoietin-related protein associated with adipose differentiation.
), and ANGPTL8 (
13Lipasin, a novel nutritionally-regulated liver-enriched factor that regulates serum triglyceride levels.
,
14- Quagliarini F.
- Wang Y.
- Kozlitina J.
- Grishin N.V.
- Hyde R.
- Boerwinkle E.
- Valenzuela D.M.
- Murphy A.J.
- Cohen J.C.
- Hobbs H.H.
Atypical angiopoietin-like protein that regulates ANGPTL3.
,
15- Ren G.
- Kim J.Y.
- Smas C.M.
Identification of RIFL, a novel adipocyte-enriched insulin target gene with a role in lipid metabolism.
).
ANGPTL3 is produced in the liver and cooperates with ANGPTL8 to inhibit LPL activity in peripheral tissues (
15- Ren G.
- Kim J.Y.
- Smas C.M.
Identification of RIFL, a novel adipocyte-enriched insulin target gene with a role in lipid metabolism.
,
16- Chi X.
- Britt E.C.
- Shows H.W.
- Hjelmaas A.J.
- Shetty S.K.
- Cushing E.M.
- Li W.
- Dou A.
- Zhang R.
- Davies B.S.J.
ANGPTL8 promotes the ability of ANGPTL3 to bind and inhibit lipoprotein lipase.
,
17- Haller J.F.
- Mintah I.J.
- Shihanian L.M.
- Stevis P.
- Buckler D.
- Alexa-Braun C.A.
- Kleiner S.
- Banfi S.
- Cohen J.C.
- Hobbs H.H.
- et al.
ANGPTL8 requires ANGPTL3 to inhibit lipoprotein lipase and plasma triglyceride clearance.
). The role of ANGPTL3/ANGPTL8 in LPL regulation is particularly important in the fed state (
18- Köster A.
- Chao Y.B.
- Mosior M.
- Ford A.
- Gonzalez-DeWhitt P.A.
- Hale J.E.
- Li D.
- Qiu Y.
- Fraser C.C.
- Yang D.D.
- et al.
Transgenic angiopoietin-like (Angptl)4 overexpression and targeted disruption of Angptl4 and Angptl3: regulation of triglyceride metabolism.
). By contrast, ANGPTL4 mainly plays a role in LPL regulation in the fasted state (
19- Lichtenstein L.
- Berbée J.F.P.
- Van Dijk S.J.
- Van Dijk K.W.
- Bensadoun A.
- Kema I.P.
- Voshol P.J.
- Müller M.
- Rensen P.C.N.
- Kersten S.
Angptl4 upregulates cholesterol synthesis in liver via inhibition of LPL- and HL-dependent hepatic cholesterol uptake.
). Individuals who carry an inactive variant of ANGPTL4 exhibit lower levels of circulating triglycerides and have decreased odds of developing coronary heart disease (
20- Abid K.
- Trimeche T.
- Mili D.
- Msolli M.A.
- Trabelsi I.
- Nouira S.
- Kenani A.
ANGPTL4 variants E40K and T266M are associated with lower fasting triglyceride levels and predicts cardiovascular disease risk in type 2 diabetic Tunisian population.
,
21- Dewey F.E.
- Gusarova V.
- O'Dushlaine C.
- Gottesman O.
- Trejos J.
- Hunt C.
- Van Hout C.V.
- Habegger L.
- Buckler D.
- Lai K-M. V.
- et al.
Inactivating variants in ANGPTL4 and risk of coronary artery disease.
). In mice, overexpression of ANGPTL4 potently represses LPL activity and leads to hypertriglyceridemia, whereas deletion of ANGPTL4 stimulates LPL activity and drastically reduces plasma triglyceride levels (
18- Köster A.
- Chao Y.B.
- Mosior M.
- Ford A.
- Gonzalez-DeWhitt P.A.
- Hale J.E.
- Li D.
- Qiu Y.
- Fraser C.C.
- Yang D.D.
- et al.
Transgenic angiopoietin-like (Angptl)4 overexpression and targeted disruption of Angptl4 and Angptl3: regulation of triglyceride metabolism.
,
22- Mandard S.
- Zandbergen F.
- Van Straten E.
- Wahli W.
- Kuipers F.
- Müller M.
- Kersten S.
The fasting-induced adipose factor/angiopoietin-like protein 4 is physically associated with lipoproteins and governs plasma lipid levels and adiposity.
). In contrast to ANGPTL3, which functions as an endocrine factor, ANGPTL4 likely serves as a local regulator of LPL in tissues where LPL and ANGPTL4 are coproduced (
23Regulation of lipid metabolism by angiopoietin-like proteins.
). Inhibition of LPL is mediated by the N-terminal coiled-coiled domain of ANGPTL4, causing the unfolding and inactivation of LPL, which may be accompanied by a change in the aggregation state of LPL (
19- Lichtenstein L.
- Berbée J.F.P.
- Van Dijk S.J.
- Van Dijk K.W.
- Bensadoun A.
- Kema I.P.
- Voshol P.J.
- Müller M.
- Rensen P.C.N.
- Kersten S.
Angptl4 upregulates cholesterol synthesis in liver via inhibition of LPL- and HL-dependent hepatic cholesterol uptake.
,
24- Mysling S.
- Kristensen K.K.
- Larsson M.
- Kovrov O.
- Bensadouen A.
- Jørgensen T.J.D.
- Olivecrona G.
- Young S.G.
- Ploug M.
The angiopoietin-like protein angptl4 catalyzes unfolding of the hydrolase domain in lipoprotein lipase and the endothelial membrane protein gpihbp1 counteracts this unfolding.
,
25- Sukonina V.
- Lookene A.
- Olivecrona T.
- Olivecrona G.
Angiopoietin-like protein 4 converts lipoprotein lipase to inactive monomers and modulates lipase activity in adipose tissue.
). In adipocytes, ANGPTL4 promotes the cleavage and subsequent degradation of LPL, thereby preventing the delivery of LPL to the endothelial surface (
26- Dijk W.
- Ruppert P.M.M.
- Oost L.J.
- Kersten S.
Angiopoietin-like 4 promotes the intracellular cleavage of lipoprotein lipase by PCSK3/furin in adipocytes.
,
27- Dijk W.
- Beigneux A.P.
- Larsson M.
- Bensadoun A.
- Young S.G.
- Kersten S.
Angiopoietin-like 4 (ANGPTL4) promotes intracellular degradation of lipoprotein lipase in adipocytes.
). The important role of ANGPTL4 in governing plasma lipid levels in humans has made ANGPTL4 an attractive therapeutic target for correcting dyslipidemia and associated cardiovascular disorders.
However, we and others have shown that disabling ANGPTL4, via monoclonal antibody-mediated or genetic inactivation, leads to a highly pro-inflammatory and ultimately lethal phenotype in mice fed a high saturated fat diet (
28- Desai U.
- Lee E.C.
- Chung K.
- Gao C.
- Gay J.
- Key B.
- Hansen G.
- Machajewski D.
- Platt K.A.
- Sands A.T.
- et al.
Lipid-lowering effects of anti-angiopoietin-like 4 antibody recapitulate the lipid phenotype found in angiopoietin-like 4 knockout mice.
,
29- Lichtenstein L.
- Mattijssen F.
- de Wit N.J.
- Georgiadi A.
- Hooiveld G.J.
- van der Meer R.
- He Y.
- Qi L.
- Köster A.
- Tamsma J.T.
- et al.
Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages.
). This marked phenotype includes mesenteric lymphadenopathy, characterized by the presence of lipid-laden Touton giant cells in the mesenteric lymph nodes, as well as fibrinopurulent peritonitis, chylous ascites, and marked elevation of acute-phase proteins in plasma, such as serum amyloid A (SAA) and haptoglobin (
29- Lichtenstein L.
- Mattijssen F.
- de Wit N.J.
- Georgiadi A.
- Hooiveld G.J.
- van der Meer R.
- He Y.
- Qi L.
- Köster A.
- Tamsma J.T.
- et al.
Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages.
,
30- Oteng A-B.
- Bhattacharya A.
- Brodesser S.
- Qi L.
- Tan N.S.
- Kersten S.
Feeding Angptl4- / - mice trans fat promotes foam cell formation in mesenteric lymph nodes without leading to ascites.
). Accumulation of lipids in mesenteric lymph nodes was also observed in several female monkeys treated with an anti-ANGPTL4 antibody (
21- Dewey F.E.
- Gusarova V.
- O'Dushlaine C.
- Gottesman O.
- Trejos J.
- Hunt C.
- Van Hout C.V.
- Habegger L.
- Buckler D.
- Lai K-M. V.
- et al.
Inactivating variants in ANGPTL4 and risk of coronary artery disease.
). Although there is so far no evidence pointing to the occurrence of abdominal lymphadenopathy in humans homozygous for an inactive ANGPTL4 variant (
21- Dewey F.E.
- Gusarova V.
- O'Dushlaine C.
- Gottesman O.
- Trejos J.
- Hunt C.
- Van Hout C.V.
- Habegger L.
- Buckler D.
- Lai K-M. V.
- et al.
Inactivating variants in ANGPTL4 and risk of coronary artery disease.
), currently the therapeutic prospects of whole-body ANGPTL4 inactivation are not very favorable.
MATERIALS AND METHODS
Animal studies
Animal studies were performed in male purebred WT,
Angptl4hyp, and
Angptl4−/− mice. All mice were on the same C57Bl/6 background strain. The
Angptl4hyp mice were a kind donation from Dr. Nguan Soon Tan (Nanyang Technological University, Singapore). The
Angptl4hyp mice can also be classified as
Angptl4 knockout first mice (
32- Skarnes W.C.
- Rosen B.
- West A.P.
- Koutsourakis M.
- Bushell W.
- Iyer V.
- Mujica A.O.
- Thomas M.
- Harrow J.
- Cox T.
- et al.
A conditional knockout resource for the genome-wide study of mouse gene function.
), which upon stepwise crossing with mice expressing flippase recombinase and Cre recombinase can be used to generate a tissue-specific
Angptl4 knockout mouse model (
33- Singh A.K.
- Aryal B.
- Chaube B.
- Rotllan N.
- Varela L.
- Horvath T.L.
- Suárez Y.
- Fernández-Hernando C.
Brown adipose tissue derived ANGPTL4 controls glucose and lipid metabolism and regulates thermogenesis.
,
34- Aryal B.
- Singh A.K.
- Zhang X.
- Varela L.
- Rotllan N.
- Goedeke L.
- Chaube B.
- Camporez J-P.
- Vatner D.F.
- Horvath T.L.
- et al.
Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis.
). The sequence of the
Angptl4 construct is available online (
https://www.i-dcc.org/imits/targ_rep/alleles/5215/escell-clone-genbank-file.)
The animal studies were all carried out at the Centre for Small Animals, which is part of the Centralized Facilities for Animal Research at Wageningen University and Research (CARUS), and were approved by the Local Animal Ethics Committee of Wageningen University (AVD104002015236: 2016.W-0093.005, 2016.W-0093.007). Mice were maintained at 21°C and kept on a regular day-night cycle (lights on from 6:00 AM to 6:00 PM).
High fat diet intervention.
Eleven- to 16-week-old mice (10 WT, 11 Angptl4hyp, and 8 Angptl4−/−) were fed a high fat diet containing 45 energy percent as triglycerides (D12451; Research Diets Inc., New Brunswick, NJ). The high fat feeding of the three groups of mice was not done in parallel. The Angptl4hyp mice were run separately from the WT and Angptl4−/− mice. After weeks 2 and 4 of the intervention, blood samples were collected to measure plasma SAA. After 20 weeks, blood was collected via orbital puncture under isoflurane anesthesia. Immediately thereafter, the mice were euthanized by cervical dislocation. Tissues were excised and immediately frozen in liquid nitrogen followed by storage at −80°C.
Fasting intervention.
Mice were fed a standard chow diet after weaning. Mice were either kept on chow or fasted for 24 h and euthanized between 9:00 and 11:00 AM. Blood was collected via orbital puncture under isoflurane anesthesia. Immediately thereafter, the mice were euthanized by cervical dislocation. Tissues were excised and immediately frozen in liquid nitrogen followed by storage at −80°C.
Intraperitoneal glucose tolerance test.
After 18 weeks of high fat diet, the mice were fasted for 5 h prior to the glucose tolerance test. The mice were injected intraperitoneally with glucose (1 g/kg body weight) (Baxter, Deerfield, IL). Blood samples from tail vein bleeding were tested for glucose levels at different time points following glucose injection using a GLUCOFIX Tech glucometer and glucose sensor test strips (Menarini Diagnostics, Valkenswaard, The Netherlands).
Histology
H&E staining was performed on the mesenteric lymph nodes. During the mouse sections, lymph nodes were isolated into plastic cassettes and immediately fixated in 4% paraformaldehyde. The tissues were processed and embedded into paraffin blocks. Thin sections of the blocks were made at 5 μm using a microtome and placed onto Superfrost glass slides followed by overnight incubation at 37°C. The tissues were stained in Mayer hematoxylin solution for 10 min and in eosin for 10 s at room temperature with intermediate washings in ethanol. The tissues were allowed to dry at room temperature followed by imaging using a light microscope.
Quantification of plasma parameters
Blood samples were collected into EDTA-coated tubes and centrifuged at 4°C for 15 min at 12,879 g. Plasma was collected and stored at −80°C. ELISA kits were used to measure plasma serum amyloid (SAA) (Tridelta Development Ltd., Ireland) and haptoglobin (Abcam, Cambridge, UK) according to manufacturer's protocol. Measurement of plasma levels of triglycerides, NEFAs, and glycerol were performed using kits from HUMAN Diagnostics (Wiesbaden, Germany) according to manufacturer's protocol. Plasma levels of cholesterol and glucose were also measured using kits from Diasys Diagnostics Systems (Holzheim, Germany) according to manufacturer's protocol.
LPL activity measurements
LPL activity levels in epididymal white adipose tissues were measured in triplicate with a [
3H]triolein-labeled lipid emulsion as previously described (
35- Kroupa O.
- Vorrsjö E.
- Stienstra R.
- Mattijssen F.
- Nilsson S.K.
- Kersten S.
- Olivecrona G.
- Olivecrona T.
Linking nutritional regulation of Angptl4, Gpihbp1, and Lmf1 to lipoprotein lipase activity in rodent adipose tissue.
). Protein contents in homogenates of adipose tissue were measured using Markwell's modified Lowry method (
36- Markwell M.A.K.
- Haas S.M.
- Bieber L.L.
- Tolbert N.E.
A modification of the Lowry procedure to simplify protein determination in membrane and lipoprotein samples.
).
Cell culture
Bone marrow cells were isolated from femurs of WT or Angptl4−/− mice following standard protocol and differentiated into macrophages [bone marrow-derived macrophages (BMDMs)] in 6–8 days in DMEM (Lonza, Verviers, Belgium) containing 10% FBS and 1% penicillin/streptomycin (PS) supplemented with 20% L929-conditioned medium. After 6–8 days, nonadherent cells were removed and adherent cells were washed and plated in 6-, 12-, or 48-well plates in DMEM/FBS/PS + 5% L929-conditioned medium. After 24 h, the cells were washed with PBS and treated. Peritoneal macrophages were obtained by infusion and subsequent collection of ice-cold PBS from the abdominal cavity and frozen directly or brought into culture in DMEM containing 10% FBS and 1% PS. RAW264.7 macrophages were cultured in DMEM containing 10% FBS and 1% PS. All cells were maintained in a humidified incubator at 37°C with 5% CO2.
Inguinal white adipose tissue from three or four WT,
Angptl4hyp, and
Angptl4−/− mice was collected and placed in DMEM supplemented with 1% PS and 1% BSA (BSA; Sigma-Aldrich). Material was minced with scissors and digested in collagenase-containing medium [DMEM with 3.2 mM CaCl
2, 1.5 mg/ml collagenase type II (C6885; Sigma-Aldrich), 10% FBS, 0.5% BSA, and 15 mM HEPES] for 1 h at 37°C with occasional vortexing. Cells were filtered through a 100 μm cell strainer (Falcon) to remove remaining cell clumps and lymph nodes. The cell suspension subsequently was centrifuged at 229
g for 10 min and the pellet was resuspended in erythrocyte lysis buffer (155 mM NH
4Cl, 12 mM NaHCO
3, 0.1 mM EDTA). Upon incubation for 2 min at room temperature, cells were centrifuged at 129
g for 5 min and the pelleted cells were resuspended in DMEM + 10% FBS + 1% PS and plated. Upon confluence, the cells were differentiated according to the protocol as described previously (
14- Quagliarini F.
- Wang Y.
- Kozlitina J.
- Grishin N.V.
- Hyde R.
- Boerwinkle E.
- Valenzuela D.M.
- Murphy A.J.
- Cohen J.C.
- Hobbs H.H.
Atypical angiopoietin-like protein that regulates ANGPTL3.
,
15- Ren G.
- Kim J.Y.
- Smas C.M.
Identification of RIFL, a novel adipocyte-enriched insulin target gene with a role in lipid metabolism.
). Briefly, confluent cells in the stromal vascular fraction were plated in 1:1 surface ratio, and differentiation was induced 2 days afterwards by switching to a differentiation induction cocktail (DMEM containing 10% FBS, 1% PS, 0.5 mM isobutylmethylxanthine, 1 μM dexamethasone, 7 μg/ml insulin, and 1 μM rosiglitazone) for 3 days. Subsequently, cells were maintained in DMEM supplemented with 10% FBS, 1% PS, and 7 μg/ml insulin for 3–6 days and switched to DMEM with 10% FBS and 1% PS for 3 days. The average rate of differentiation was at least 80% as determined by eye.
Cell culture experiments and chemical treatments
BMDMs and peritoneal macrophages were exposed to 0.5 mM intralipid or 0.5 mM oleic acid (Sigma-Aldrich) for 6 h in DMEM/PS media. The oleic acid was conjugated to BSA at a ratio of 2:1 (BSA:oleic acid). BMDMs were also treated with synthetic PPAR agonists (1 μM Wy14643, 1 μM rosiglitazone, 1 μM L165041, 1 μM GW501516) or vehicle control for 6 h. All PPAR agonists were obtained from Sigma-Aldrich. Peritoneal macrophages and RAW264.7 cells were incubated for 6 h with lymph (final triglyceride concentration of 2 mM, which was collected from rats provided with palm-oil based high fat diet overnight). BMDMs were co-incubated for 6 h with 10 μM orlistat (Sigma-Aldrich) and 0.5 mM intralipid. RAW264.7 macrophages treated with 0.5 mM intralipid or lymph were incubated with or without 0.5 μg/ml recombinant ANGPTL4 (R&D Systems, Abingdon, UK). In a separate experiment, BMDMs of WT and Angptl4−/− mice were exposed to the ER to Golgi transport inhibitors, monensin (10 μM, 3 h; Cayman Chemicals) and Brefeldin A (5 μg/ml, 4 h; Sigma-Aldrich), proteasomal degradation inhibitor, MG132 (40 μM, 4 h; Sigma-Aldrich), and lysosomal degradation inhibitors, e64d (20 μM, 24 h; Sigma-Aldrich) and leupeptin (5 μM, 16 h; Cayman Chemicals).
Oil Red O staining
Oil Red O staining was performed in RAW264.7 cells incubated with intralipid or lymph in the presence or absence of recombinant ANGPTL4. A stock of Oil Red O (Sigma) was prepared by dissolving 0.5 g in 500 ml of isopropanol. Working concentrations were made by dissolving stock concentrations with water (3 stock:2 water) and filtered. Treated cells were washed twice with PBS followed by fixation with 4% formalin for 30 min. The cells were then washed twice with PBS and incubated with Oil Red O dye for 20 min. The stained cells were washed three times with ddH2O after which cells were visualized under a light microscope and pictures taken.
Bodipy staining
For visualization of lipid droplets, BMDMs of WT and Angptl4−/− mice were washed with PBS, fixated in 4% formalin, and subsequently stained with 1 ng/ml Bodipy 493/503 (Thermo Fisher Scientific, Landsmeer, The Netherlands).
RNA isolation and quantitative real-time PCR
Total RNA was isolated from tissues and cells by homogenizing in TRIzol (Thermo Fisher Scientific) either with a QIagen Tissue Lyser II (Qiagen, Venlo, The Netherlands) or by pipetting up and down. Reverse transcription was performed using the iScript™ cDNA synthesis kit (Bio-Rad) according to the manufacturer's protocol. Quantitative PCR amplifications were done on a CFX384 real-time PCR platform (Bio-Rad) with the SensiMix PCR mix from Bioline (GC Biotech, Alphen aan de Rijn, The Netherlands). Primer sequences of genes are provided in
supplemental Table S1. Gene expression values were normalized to the housekeeping gene,
36b4 (
Rplp0, ribosomal protein lateral stalk subunit P0).
Microarray analysis
RNeasy mini columns (Qiagen) were used to isolate RNA from mouse peritoneal macrophages that were incubated for 6 h with 0.5 mM intralipid in the presence or absence of 2.5 μg/ml recombinant ANGPTL4. RNA quality was verified on an Agilent 2100 bioanalyzer (Agilent Technologies, Amsterdam, The Netherlands) using 6000 Nano chips according to the manufacturer's instructions. RNA was considered suitable for array hybridization only if the RNA integrity number exceeded 8.0. RNA from three samples per group was pooled for microarray analysis. One hundred nanograms of RNA were used for Whole Transcript cDNA synthesis (Affymetrix, Santa Clara, CA). Hybridization, washing, and scanning of Affymetrix GeneChip Mouse Gene 1.0 ST arrays were carried out according to standard Affymetrix protocols. Scans of the Affymetrix arrays were processed using packages from the Bioconductor project. Arrays were normalized using the Robust Multi-array Average method (
37- Bolstad B.M.
- Irizarry R.A.
- Astrand M.
- Speed T.P.
A comparison of normalization methods for high density oligonucleotide array data based on variance and bias.
,
38- Irizarry R.A.
- Hobbs B.
- Collin F.
- Beazer-Barclay Y.D.
- Antonellis K.J.
- Scherf U.
- Speed T.P.
Exploration, normalization, and summaries of high density oligonucleotide array probe level data.
). Probe sets were defined by assigning probes to unique gene identifiers, e.g., Entrez ID (
39- Dai M.
- Wang P.
- Boyd A.D.
- Kostov G.
- Athey B.
- Jones E.G.
- Bunney W.E.
- Myers R.M.
- Speed T.P.
- Akil H.
- et al.
Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data.
). Changes in gene expression were calculated as signal log ratios between treatment and control. These ratios were used to create heat maps within Excel. The CEL files were deposited in Gene Expression Omnibus (accession number GSE136240).
Affymetrix GeneChip analysis was carried out on WT mouse BMDMs incubated with oleate (250 μM) for 5 h (
40- Robblee M.M.
- Kim C.C.
- Abate J.P.
- Valdearcos M.
- Sandlund K.L.M.
- Shenoy M.K.
- Volmer R.
- Iwawaki T.
- Koliwad S.K.
Saturated fatty acids engage an IRE1α-dependent pathway to activate the NLRP3 inflammasome in myeloid cells.
). CEL files were downloaded from the internet via Gene Expression Omnibus (GSE77104) and processed as described above.
Extracellular flux analysis
Real-time oxygen consumption rates (OCRs) of BMDMs from WT and Angptl4−/− mice were assessed using XF-96 extracellular flux analyzer (Seahorse Bioscience, Santa Clara, CA). Basal metabolic rates of BMDMs seeded in quintuplicate were determined during three consecutive measurements in unbuffered Seahorse medium [8.3 g DMEM powder, 0.016 g phenol red, and 1.85 g NaCl in 1 liter milli-Q (pH 7.4) at 37°C, sterile-filtered] containing 25 mM glucose and 2 mM L-glutamine. Measurements were performed after 6 h treatment with intralipid (0.5 mM) in the presence or absence of recombinant ANGPTL4 (0.5 ug/ml). After basal measurements, three consecutive measurements were made following addition of 1.5 μM oligomycin, 1.5 μM carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), and combination of 2 μM antimycin A and 1 μM rotenone. Pyruvate (1 mM) was added together with FCCP to fuel maximal respiration. All compounds used during the Seahorse runs were acquired from Merck. Signals were normalized to relative DNA content in the wells using the Quanti-iT™ dsDNA assay kit (Thermo Fisher Scientific).
Western immunoblotting
To isolate protein, mouse fat pads, differentiated primary adipocytes, primary macrophages, and whole adipose tissues were lysed in RIPA lysis and extraction buffer [25 mM Tris-HCl (pH 7.6), 150 mM NaCl, 1% Nonidet P-40, and 0.1% SDS; Thermo Fisher Scientific] supplemented with protease and phosphatase inhibitors (Roche Diagnostics, Almere, The Netherlands). Following homogenization, lysates were placed on ice for 30 min and centrifuged two or three times at 12,879
g for 10 min at 4°C to remove fat and cell debris. Concentration of protein lysates was determined using a bicinchoninic acid assay (Thermo Fisher Scientific). For assessment of LPL release, BMDMs were treated for 20 min with 10 IU/ml heparin (#012866-08; LEO Pharma). For assessment of glycosylation of LPL, 2–30 μg of proteins were digested with endoglycosidase H (EndoH) (New England BioLabs) according to manufacturer's protocol. Protein lysates (10–30 μg of protein per lane) were loaded onto 8–16% or 10% Criterion gels (Bio-Rad, Veenendaal, The Netherlands). Next, proteins were transferred onto a polyvinylidene difluoride membrane using the Transblot Turbo system (Bio-Rad). Membranes were probed with a goat anti-mouse LPL antibody (
41- Weinstein M.M.
- Yin L.
- Beigneux A.P.
- Davies B.S.
- Gin P.
- Estrada K.
- Melford K.
- Bishop J.R.
- Esko J.D.
- Dallinga-Thie G.M.
- et al.
Abnormal patterns of lipoprotein lipase release into the plasma in GPIHBP1-deficient mice.
), a rabbit anti-mouse HSP90 antibody (#4874S; Cell Signaling), a rat anti-mouse ANGPTL4 antibody (Kairos 142-2; Adipogen), and a rabbit anti-mouse ANGPTL4 antibody (#742, home made) (
42- Mandard S.
- Zandbergen F.
- Nguan S.T.
- Escher P.
- Patsouris D.
- Koenig W.
- Kleemann R.
- Bakker A.
- Veenman F.
- Wahli W.
- et al.
The direct peroxisome proliferator-activated receptor target fasting-induced adipose factor (FIAF/PGAR/ANGPTL4) is present in blood plasma as a truncated protein that is increased by fenofibrate treatment.
) at 1:5,000 (LPL), 1:2,000 (HSP90), or 1:1,000 (ANGPTL4) dilutions. Blocking and incubation of primary and secondary antibodies were done in TBS (pH 7.5) plus 0.1% Tween 20 (TBS-T) and 5% (w/v) skimmed milk at 1:5,000. In between, membranes were washed in TBS-T. Quantification was performed with the ChemiDoc MP system (Bio-Rad) and Clarity ECL substrate (Bio-Rad). Equal loading of medium samples was verified with HSP90.
Statistical analysis
Statistical analyses were performed using one-way ANOVA followed by Tukey HSD test or by Student's t-test. Data are presented as mean ± SEM (animal experiments) or mean ± SD (cell culture studies). P < 0.05 was considered statistically significant.
DISCUSSION
In this work, we addressed two independent questions relevant to the mesenteric lymphadenopathy phenotype in Angptl4−/− mice fed a high fat diet: 1) what is the influence of endogenous ANGPTL4 on lipid uptake and utilization in macrophages; and 2) what is the role of N-terminal ANGPTL4 in the development of mesenteric lymphadenopathy, inflammation, and ascites?
Our study confirms previous data indicating that
Angptl4 expression in macrophages is highly induced by lipids (
29- Lichtenstein L.
- Mattijssen F.
- de Wit N.J.
- Georgiadi A.
- Hooiveld G.J.
- van der Meer R.
- He Y.
- Qi L.
- Köster A.
- Tamsma J.T.
- et al.
Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages.
,
45- Georgiadi A.
- Wang Y.
- Stienstra R.
- Tjeerdema N.
- Janssen A.
- Stalenhoef A.
- van der Vliet J.A.
- de Roos A.
- Tamsma J.T.
- Smit J.W.
- et al.
Overexpression of angiopoietin-like protein 4 protects against atherosclerosis development.
,
46- Aryal B.
- Rotllan N.
- Araldi E.
- Ramírez C.M.
- He S.
- Chousterman B.G.
- Fenn A.M.
- Wanschel A.
- Madrigal-Matute J.
- Warrier N.
- et al.
ANGPTL4 deficiency in haematopoietic cells promotes monocyte expansion and atherosclerosis progression.
). This induction is likely mediated by PPARδ and/or PPARγ, as shown by the marked upregulation of
Angptl4 mRNA by PPARδ and PPARγ agonists in BMDMs. Importantly, the present study shows that not only external ANGPTL4 but also endogenously produced ANGPTL4 inhibits lipid uptake in macrophages. The elevated lipid uptake in
Angptl4−/− macrophages increases the expression of various lipid-sensitive genes involved in inflammation and ER stress, such as
Hilpda,
Ddit3,
Ptgs2, and
Cxcl2. Interestingly, the elevated lipid uptake in
Angptl4−/− macrophages was accompanied by increased mitochondrial respiration, suggesting that enhanced lipid uptake may stimulate cellular respiration. As the effect on cellular respiration was observed for endogenous and not exogenous ANGPTL4, it might reflect an intracellular mechanism of action of ANGPTL4. Together, our findings demonstrate an important role of ANGPTL4 in regulating lipid uptake and utilization in macrophages.
It is well-established that ANGPTL4 inactivates LPL by promoting LPL unfolding (
24- Mysling S.
- Kristensen K.K.
- Larsson M.
- Kovrov O.
- Bensadouen A.
- Jørgensen T.J.D.
- Olivecrona G.
- Young S.G.
- Ploug M.
The angiopoietin-like protein angptl4 catalyzes unfolding of the hydrolase domain in lipoprotein lipase and the endothelial membrane protein gpihbp1 counteracts this unfolding.
,
25- Sukonina V.
- Lookene A.
- Olivecrona T.
- Olivecrona G.
Angiopoietin-like protein 4 converts lipoprotein lipase to inactive monomers and modulates lipase activity in adipose tissue.
). Recently, we found that ANGPTL4 has a local role in adipocytes by promoting the intracellular cleavage and subsequent degradation of LPL (
26- Dijk W.
- Ruppert P.M.M.
- Oost L.J.
- Kersten S.
Angiopoietin-like 4 promotes the intracellular cleavage of lipoprotein lipase by PCSK3/furin in adipocytes.
,
27- Dijk W.
- Beigneux A.P.
- Larsson M.
- Bensadoun A.
- Young S.G.
- Kersten S.
Angiopoietin-like 4 (ANGPTL4) promotes intracellular degradation of lipoprotein lipase in adipocytes.
). In stark contrast to adipocytes, ANGPTL4 deficiency in macrophages did not lead to the accumulation of full-length rapidly releasable LPL, while the cleaved N-terminal LPL portion was not detectable in our hands. The lack of effect of ANGPTL4 on LPL protein levels in macrophages may be because macrophage LPL is not degraded via the lysosomal pathway but via the proteasomal pathway. The difference in the LPL degradation pathway and in the effect of ANGPTL4 on LPL levels between adipocytes and macrophages could be connected to how LPL is presented on the cell surface. Specifically, it is conceivable that LPL on the surface of macrophages is not internalized for degradation, unlike LPL in adipocytes, and that ANGPTL4 only causes the unfolding and inactivation of LPL after both proteins have been secreted (
47- Cupp M.
- Bensadoun A.
- Melford K.
Heparin decreases the degradation rate of lipoprotein lipase in adipocytes.
,
48- He P.P.
- Jiang T.
- OuYang X.P.
- Liang Y.Q.
- Zou J.Q.
- Wang Y.
- Shen Q.Q.
- Liao L.
- Zheng X.L.
Lipoprotein lipase: biosynthesis, regulatory factors, and its role in atherosclerosis and other diseases.
). Overall, these findings suggest that the location and cellular mechanism of LPL inactivation by ANGPTL4 may be different in different cells.
Previously, we showed that
Angptl4−/− mice fed a diet rich in saturated fat develop an inflammatory and ultimately lethal phenotype characterized by the formation of Touton giant cells in mesenteric lymph nodes, chylous ascites, and fibrinopurulent peritonitis (
29- Lichtenstein L.
- Mattijssen F.
- de Wit N.J.
- Georgiadi A.
- Hooiveld G.J.
- van der Meer R.
- He Y.
- Qi L.
- Köster A.
- Tamsma J.T.
- et al.
Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages.
,
30- Oteng A-B.
- Bhattacharya A.
- Brodesser S.
- Qi L.
- Tan N.S.
- Kersten S.
Feeding Angptl4- / - mice trans fat promotes foam cell formation in mesenteric lymph nodes without leading to ascites.
). By comparing
Angptl4−/− mice with an
Angptl4-hypomorphic model, here we show that a low-level expression of a truncated N-terminal ANGPTL4 does not prevent Touton giant cell formation, yet drastically mitigates the acute-phase response and the development of chylous ascites upon high fat feeding. Importantly, the development of fibrinopurulent peritonitis and the death of the animals is completely prevented by the expression of the truncated N-terminal ANGPTL4. Although the severity of the clinical phenotype was attenuated, activation of the acute-phase response and chylous ascites were still observed in a substantial number of
Angptl4hyp mice.
Strictly, we cannot fully exclude that N-terminal ANGPTL4 influences ascites independently of LPL inhibition. However, two lines of reasoning support an action of N-terminal ANGPTL4 on ascites via LPL. First, LPL protein levels in
Angptl4hyp adipocytes were lower than in
Angptl4−/− adipocytes, paralleled by a marked reduction in ascites severity. These data suggest that a low level of N-terminal ANGPTL4 decreases LPL abundance. Second, chronic injection of a monoclonal antibody that is directed against N-terminal ANGPTL4 and abolishes its ability to inhibit LPL causes ascites in mice fed a HFD (
28- Desai U.
- Lee E.C.
- Chung K.
- Gao C.
- Gay J.
- Key B.
- Hansen G.
- Machajewski D.
- Platt K.A.
- Sands A.T.
- et al.
Lipid-lowering effects of anti-angiopoietin-like 4 antibody recapitulate the lipid phenotype found in angiopoietin-like 4 knockout mice.
). Collectively, we favor the notion that the attenuation of the ascites phenotype in
Angptl4hyp mice compared with
Angptl4−/− mice is related to reduced LPL activity via N-terminal ANGPTL4 and, accordingly, that the ascites in
Angptl4−/− mice fed a high fat diet is directly connected to enhanced LPL activity.
Our observation that partial deficiency of ANGPTL4 leads to undesirable clinical consequences in mice suggests that therapeutic approaches that only partially inactivate ANGPTL4 and/or are aimed at whole-body inactivation of ANGPTL4 may still carry the risk of major side effects in humans. By contrast, tissue-specific inactivation, in particular adipose tissue-specific and possibly liver-specific inactivation, of ANGPTL4 holds considerably more promise for improving dyslipidemia and reducing coronary artery disease risk without leading to deleterious side-effects (
34- Aryal B.
- Singh A.K.
- Zhang X.
- Varela L.
- Rotllan N.
- Goedeke L.
- Chaube B.
- Camporez J-P.
- Vatner D.F.
- Horvath T.L.
- et al.
Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis.
).
In line with a predominant role of ANGPTL4 in lipid metabolism during fasting, plasma levels of triglycerides, cholesterol, and NEFAs were lower in
Angptl4−/− mice compared with WT mice specifically in the fasted but not the fed state. Interestingly, in the fasted state, plasma triglyceride, cholesterol, and NEFA levels were similar in
Angptl4hyp and
Angptl4−/− mice, which was accompanied by similar levels of LPL protein in the adipose tissue. Accordingly, the expression of low levels of N-terminal ANGPTL4 does not influence LPL protein levels in the fasted state. By contrast, in the fed state, LPL protein levels in adipose tissue were lower in
Angptl4hyp mice than in
Angptl4−/− mice, suggesting that a low level of N-terminal ANGPTL4 decreases LPL protein, likely by promoting LPL degradation. The reason why N-terminal ANGPTL4 in
Angptl4hyp mice has a more pronounced effect on LPL in the fed state than in the fasted state is unclear. Why fasting fails to alter
Angptl4 mRNA in
Angptl4hyp mice is also unclear. It can be hypothesized that it is due to a disrupted transcriptional regulation by the glucocorticoid receptor via exon 7, as this mechanism is responsible for the induction of
Angptl4 expression by fasting in WT adipose tissue (
49- Koliwad S.K.
- Kuo T.
- Shipp L.E.
- Gray N.E.
- Backhed F.
- So A.Y.L.
- Farese R.V.
- Wang J.C.
Angiopoietin-like 4 (ANGPTL4, fasting-induced adipose factor) is a direct glucocorticoid receptor target and participates in glucocorticoid-regulated triglyceride metabolism.
,
50- Gray N.E.
- Lam L.N.
- Yang K.
- Zhou A.Y.
- Koliwad S.
- Wang J.C.
Angiopoietin-like 4 (Angptl4) protein is a physiological mediator of intracellular lipolysis in murine adipocytes.
). Despite the minimal effect of fasting on levels of N-terminal ANGPTL4 in
Angptl4hyp mice, LPL protein levels increased, suggesting an additional regulatory mechanism of LPL protein.
In addition to regulating plasma lipid levels, recent reports also suggest a role of ANGPTL4 in glucose homeostasis (
51Can targeting ANGPTL proteins improve glucose tolerance?.
). Using different models, ANGPTL4 deficiency in mice was found to lead to improved glucose tolerance. Consistent with the study by Gusarova et al. (
52- Gusarova V.
- O'Dushlaine C.
- Teslovich T.M.
- Benotti P.N.
- Mirshahi T.
- Gottesman O.
- Van Hout C.V.
- Murray M.F.
- Mahajan A.
- Nielsen J.B.
- et al.
Genetic inactivation of ANGPTL4 improves glucose homeostasis and is associated with reduced risk of diabetes.
), we found that
Angptl4−/− mice chronically fed a high fat diet have drastically lower basal plasma glucose levels and improved glucose tolerance. However, these mice have numerous clinical abnormalities, which prohibit any conclusion on the direct effect of ANGPTL4 deficiency on glucose homeostasis.
Angptl4hyp mice fed a high fat diet showed a much milder clinical phenotype, concomitant with a more modest yet statistically significant improvement in glucose tolerance; yet acute-phase protein levels were still elevated compared with WT mice. This observation again makes it difficult to derive a solid conclusion on the direct effect of ANGPTL4 deficiency on glucose homeostasis. We previously found that
Angptl4−/− mice fed a diet high in unsaturated fat, which does not exhibit any clinical complications, were more glucose tolerant than WT mice fed the same diet (
53- Janssen A.W.F.
- Katiraei S.
- Bartosinska B.
- Eberhard D.
- Van Dijk K.W.
- Kersten S.
Loss of angiopoietin-like 4 (ANGPTL4) in mice with diet-induced obesity uncouples visceral obesity from glucose intolerance partly via the gut microbiota.
). In addition, it was shown that adipocyte-specific ANGPTL4-deficient mice have markedly improved glucose and insulin tolerance after 1 month and to a lesser extent after five months of high fat feeding, which was suggested to be due to LPL-mediated redistribution of ectopic fat stores to adipose tissue (
34- Aryal B.
- Singh A.K.
- Zhang X.
- Varela L.
- Rotllan N.
- Goedeke L.
- Chaube B.
- Camporez J-P.
- Vatner D.F.
- Horvath T.L.
- et al.
Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis.
). While growing evidence thus connects ANGPTL4 with glucose homeostasis, further studies are necessary to delineate potential mechanisms.
Another interesting question concerns the significance of the lipid-laden Touton giant cells for the onset of the debilitating side effects after high fat feeding. A causal role had been previously questioned by the observation that
Angptl4−/− mice fed a diet high in
trans fatty acids develop Touton cells but do not show elevated systemic inflammation or ascites and survive the intervention (
30- Oteng A-B.
- Bhattacharya A.
- Brodesser S.
- Qi L.
- Tan N.S.
- Kersten S.
Feeding Angptl4- / - mice trans fat promotes foam cell formation in mesenteric lymph nodes without leading to ascites.
). Here, we found that
Angptl4hyp mice fed a regular high fat diet, despite showing a much milder inflammation and ascites than
Angptl4−/− mice, carried similar numbers of Touton giant cells in their mesenteric lymph nodes. These data reinforce the notion that the formation of lipid-laden Touton giant cells is uncoupled from activation of an acute-phase response and chylous ascites (
30- Oteng A-B.
- Bhattacharya A.
- Brodesser S.
- Qi L.
- Tan N.S.
- Kersten S.
Feeding Angptl4- / - mice trans fat promotes foam cell formation in mesenteric lymph nodes without leading to ascites.
).
Two previous publications have used the
Angptl4hyp mice to generate floxed
Angptl4 mice, which in turn were used to generate adipocyte- or brown fat-specific ANGPTL4-deficient mice (
33- Singh A.K.
- Aryal B.
- Chaube B.
- Rotllan N.
- Varela L.
- Horvath T.L.
- Suárez Y.
- Fernández-Hernando C.
Brown adipose tissue derived ANGPTL4 controls glucose and lipid metabolism and regulates thermogenesis.
,
34- Aryal B.
- Singh A.K.
- Zhang X.
- Varela L.
- Rotllan N.
- Goedeke L.
- Chaube B.
- Camporez J-P.
- Vatner D.F.
- Horvath T.L.
- et al.
Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis.
). Deficiency of ANGPTL4 was shown by dramatically reduced qPCR-based amplification of introns 4–5, which covers the region removed by Cre-mediated excision. No information was presented on ANGPTL4 protein levels. Based on our results and calculations, it is possible that in both mouse models there is still significant expression of a truncated N-terminal ANGPTL4 protein derived from exons 1–3 containing the LPL-inhibitory domain. It can be argued that this truncated ANGPTL4 might influence the metabolic phenotype.
In conclusion, we found that ANGPTL4 deficiency increased lipid uptake and respiration in macrophages without affecting LPL protein levels. Furthermore, in comparison to Angptl4−/− mice, mice expressing low levels of N-terminal ANGPTL4 showed a reduced acute-phase response and markedly attenuated chylous ascites following high fat feeding. These findings have significant clinical implications inasmuch as any therapeutic strategy would likely reduce but not completely inactivate ANGPTL4.
Article info
Publication history
Published online: August 13, 2019
Received in revised form:
August 13,
2019
Received:
March 21,
2019
Footnotes
This work was supported by the Graduate School Voeding, Levensmiddelentechnologie, Agro-Biotechnologie en Gezondheid (VLAG) (Wageningen University), and CVON ENERGISE Grant CVON2014-02. The authors declare that there are no conflicts of interest associated with this work.
The online version of this article (available at http://www.jlr.org) contains a supplement.
The data discussed in this publication have been deposited in NCBI's Gene Expression Omnibus (Oteng et al., 2019) and are accessible through GEO Series accession number GSE136240; (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE136240).
Abbreviations:
ANGPTLangiopoietin-like protein
BMDMbone marrow-derived macrophage
FCCPcarbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone
OCRoxygen consumption rate
PSpenicillin/streptomycin
SAAserum amyloid A
Copyright
Copyright © 2019 Oteng et al.