apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while...

10
RESOURCE ARTICLE Apoc2 loss-of-function zebrafish mutant as a genetic model of hyperlipidemia Chao Liu 1 , Keith P. Gates 2 , Longhou Fang 1, *, Marcelo J. Amar 3 , Dina A. Schneider 1 , Honglian Geng 1 , Wei Huang 1 , Jungsu Kim 1 , Jennifer Pattison 1 , Jian Zhang 4 , Joseph L. Witztum 1 , Alan T. Remaley 3 , P. Duc Dong 2 and Yury I. Miller 1, ABSTRACT Apolipoprotein C-II (APOC2) is an obligatory activator of lipoprotein lipase. Human patients with APOC2 deficiency display severe hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis. Hypertriglyceridemia is also an important risk factor for development of cardiovascular disease. Animal models to study hypertriglyceridemia are limited, with no Apoc2-knockout mouse reported. To develop a genetic model of hypertriglyceridemia, we generated an apoc2 mutant zebrafish characterized by the loss of Apoc2 function. apoc2 mutants show decreased plasma lipase activity and display chylomicronemia and severe hypertriglyceridemia, which closely resemble the phenotype observed in human patients with APOC2 deficiency. The hypertriglyceridemia in apoc2 mutants is rescued by injection of plasma from wild-type zebrafish or by injection of a human APOC2 mimetic peptide. Consistent with a previous report of a transient apoc2 knockdown, apoc2 mutant larvae have a minor delay in yolk consumption and angiogenesis. Furthermore, apoc2 mutants fed a normal diet accumulate lipid and lipid-laden macrophages in the vasculature, which resemble early events in the development of human atherosclerotic lesions. In addition, apoc2 mutant embryos show ectopic overgrowth of pancreas. Taken together, our data suggest that the apoc2 mutant zebrafish is a robust and versatile animal model to study hypertriglyceridemia and the mechanisms involved in the pathogenesis of associated human diseases. KEY WORDS: Zebrafish, Apolipoprotein C-II, APOC2, Lipoprotein lipase, Hyperlipidemia INTRODUCTION Hypertriglyceridemia is an independent risk factor for cardiovascular disease (CVD) (Do et al., 2013) and is positively associated with obesity, insulin resistance, type 2 diabetes and other metabolic syndromes (Watts et al., 2013). Hypertriglyceridemia is the result of interactions between non-genetic and genetic factors. The most common non-genetic factors causing hypertriglyceridemia are obesity, alcohol excess, lack of exercise and unhealthy diets (Miller et al., 2011; Watts et al., 2013). Genetic factors account for 50% of individual variation in plasma triglyceride (TG) levels (Goldberg et al., 2011; Miller et al., 2011; Namboodiri et al., 1985), whereas severe hypertriglyceridemia (TG>885 mg/dl) is usually caused by single mutations in the gene encoding lipoprotein lipase (LPL), or less commonly, in genes affecting proteins involved in LPL activity, such as apolipoprotein C-II (APOC2), lipase maturation factor 1 (LMF1), apolipoprotein A-V (APOA5) and GPI HDL binding protein1 (GPIHBP1) (Surendran et al., 2012). APOC2 is an obligatory co-activating factor for LPL, which is the key enzyme responsible for hydrolysis of plasma TG (Fukushima and Yamamoto, 2004; Kei et al., 2012). Patients with APOC2 or LPL deficiency show severe hypertriglyceridemia and chylomicronemia, and often manifest eruptive xanthomas, lipemia retinalis and acute and recurrent pancreatitis, which can be lethal (Breckenridge et al., 1978; Cox et al., 1978; Ewald et al., 2009; Goldberg and Merkel, 2001; Miller et al., 2011; Scherer et al., 2014; Watts et al., 2013). Current genetic animal models to study hyperlipidemia include mice with a conditional deficiency in LPL, deficiency of GPIHBP1, or with overexpression of human APOC2 and APOC3 (Ebara et al., 1997; Goulbourne et al., 2014; Shachter et al., 1994; Weinstein et al., 2010). There are several systemic or tissue specific Lpl-knockout mouse models that have been developed (Garcia-Arcos et al., 2013; Merkel et al., 1998; Trent et al., 2014; Weinstock et al., 1995). The systemic Lpl-knockout mice develop hypertriglyceridemia but die shortly after birth. The neonatal death, together with the hypertriglyceridemia, are prevented by overexpression of human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995). In contrast to extensive work conducted with Lpl-knockout mice, Apoc2-knockout mouse models have not been reported. Zebrafish is an emerging animal model to study lipid metabolism and mechanisms of human disease related to lipid abnormalities (Anderson et al., 2011; Fang et al., 2014; Holtta-Vuori et al., 2010; Levic et al., 2015; OHare et al., 2014; Stoletov et al., 2009). The advantages of using zebrafish include large progeny numbers, optical transparency of zebrafish larvae, easy genetic manipulation and cost-effective maintenance (Dooley and Zon, 2000). Importantly, genes involved in lipid and lipoprotein metabolism, such as APOB, APOE, APOA1, LDLR, APOC2, LPL, LCAT and CETP, are conserved from zebrafish to humans (Fang et al., 2014; Holtta-Vuori et al., 2010; Otis et al., 2015). Specifically, the zebrafish Lpl (NCBI Gene ID: 30354) amino acid sequence is 61.7% identical to its human ortholog, with 79.7% conserved consensus amino acids. The zebrafish full-length Apoc2 (NCBI Gene ID: 568972, molecular mass 11.2 kDa) is only 27.8% Received 2 January 2015; Accepted 29 May 2015 1 Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA. 2 Sanford Childrens Health Research Center, Programs in Genetic Disease and Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA. 3 Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, NIH, Bethesda, MD, USA. 4 State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China. *Present address: Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA. Author for correspondence ([email protected]) This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 989 © 2015. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836 Disease Models & Mechanisms

Upload: others

Post on 26-Sep-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

RESOURCE ARTICLE

Apoc2 loss-of-function zebrafish mutant as a genetic model ofhyperlipidemiaChao Liu1, Keith P. Gates2, Longhou Fang1,*, Marcelo J. Amar3, Dina A. Schneider1, HonglianGeng1,Wei Huang1,Jungsu Kim1, Jennifer Pattison1, Jian Zhang4, Joseph L. Witztum1, Alan T. Remaley3, P. Duc Dong2 andYury I. Miller1,‡

ABSTRACTApolipoprotein C-II (APOC2) is an obligatory activator of lipoproteinlipase. Human patients with APOC2 deficiency display severehypertriglyceridemia while consuming a normal diet, often manifestingxanthomas, lipemia retinalis and pancreatitis. Hypertriglyceridemiais also an important risk factor for development of cardiovasculardisease. Animal models to study hypertriglyceridemia are limited, withno Apoc2-knockout mouse reported. To develop a genetic model ofhypertriglyceridemia, we generated an apoc2 mutant zebrafishcharacterized by the loss of Apoc2 function. apoc2 mutants showdecreased plasma lipase activity and display chylomicronemia andsevere hypertriglyceridemia, which closely resemble the phenotypeobserved in human patients with APOC2 deficiency. Thehypertriglyceridemia in apoc2 mutants is rescued by injection ofplasma from wild-type zebrafish or by injection of a human APOC2mimetic peptide. Consistent with a previous report of a transient apoc2knockdown, apoc2 mutant larvae have a minor delay in yolkconsumption and angiogenesis. Furthermore, apoc2 mutants fed anormal diet accumulate lipid and lipid-laden macrophages in thevasculature, which resemble early events in the development ofhuman atherosclerotic lesions. In addition, apoc2 mutant embryosshow ectopic overgrowth of pancreas. Taken together, our data suggestthat the apoc2mutant zebrafish is a robust and versatile animalmodel tostudy hypertriglyceridemia and the mechanisms involved in thepathogenesis of associated human diseases.

KEY WORDS: Zebrafish, Apolipoprotein C-II, APOC2,Lipoprotein lipase, Hyperlipidemia

INTRODUCTIONHypertriglyceridemia is an independent risk factor for cardiovasculardisease (CVD) (Do et al., 2013) and is positively associated withobesity, insulin resistance, type 2 diabetes and other metabolic

syndromes (Watts et al., 2013). Hypertriglyceridemia is the result ofinteractions between non-genetic and genetic factors. The mostcommon non-genetic factors causing hypertriglyceridemia areobesity, alcohol excess, lack of exercise and unhealthy diets (Milleret al., 2011; Watts et al., 2013). Genetic factors account for 50% ofindividual variation in plasma triglyceride (TG) levels (Goldberget al., 2011; Miller et al., 2011; Namboodiri et al., 1985), whereassevere hypertriglyceridemia (TG>885 mg/dl) is usually caused bysingle mutations in the gene encoding lipoprotein lipase (LPL), orless commonly, in genes affecting proteins involved in LPL activity,such as apolipoprotein C-II (APOC2), lipase maturation factor 1(LMF1), apolipoprotein A-V (APOA5) and GPI HDL bindingprotein1 (GPIHBP1) (Surendran et al., 2012).

APOC2 is an obligatory co-activating factor for LPL, which is thekey enzyme responsible for hydrolysis of plasma TG (Fukushima andYamamoto, 2004; Kei et al., 2012). Patients with APOC2 or LPLdeficiency show severe hypertriglyceridemia and chylomicronemia,and often manifest eruptive xanthomas, lipemia retinalis and acuteand recurrent pancreatitis, which can be lethal (Breckenridge et al.,1978; Cox et al., 1978; Ewald et al., 2009; Goldberg and Merkel,2001; Miller et al., 2011; Scherer et al., 2014; Watts et al., 2013).Current genetic animal models to study hyperlipidemia include micewith a conditional deficiency inLPL, deficiencyofGPIHBP1, orwithoverexpression of human APOC2 and APOC3 (Ebara et al., 1997;Goulbourne et al., 2014; Shachter et al., 1994;Weinstein et al., 2010).There are several systemic or tissue specific Lpl-knockout mousemodels that have been developed (Garcia-Arcos et al., 2013; Merkelet al., 1998; Trent et al., 2014; Weinstock et al., 1995). The systemicLpl-knockout mice develop hypertriglyceridemia but die shortly afterbirth. The neonatal death, together with the hypertriglyceridemia, areprevented by overexpression of human LPL in either skeletal orcardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995). Incontrast to extensive work conducted with Lpl-knockout mice,Apoc2-knockout mouse models have not been reported.

Zebrafish is an emerging animal model to study lipid metabolismand mechanisms of human disease related to lipid abnormalities(Anderson et al., 2011; Fang et al., 2014; Holtta-Vuori et al., 2010;Levic et al., 2015; O’Hare et al., 2014; Stoletov et al., 2009). Theadvantages of using zebrafish include large progeny numbers,optical transparency of zebrafish larvae, easy genetic manipulationand cost-effective maintenance (Dooley and Zon, 2000).Importantly, genes involved in lipid and lipoprotein metabolism,such as APOB, APOE, APOA1, LDLR, APOC2, LPL, LCAT andCETP, are conserved from zebrafish to humans (Fang et al., 2014;Holtta-Vuori et al., 2010; Otis et al., 2015). Specifically, thezebrafish Lpl (NCBI Gene ID: 30354) amino acid sequence is61.7% identical to its human ortholog, with 79.7% conservedconsensus amino acids. The zebrafish full-length Apoc2 (NCBIGene ID: 568972, molecular mass 11.2 kDa) is only 27.8%Received 2 January 2015; Accepted 29 May 2015

1Division of Endocrinology and Metabolism, Department of Medicine, University ofCalifornia, San Diego, La Jolla, CA, USA. 2Sanford Children’s Health ResearchCenter, Programs in Genetic Disease and Development and Aging, and Stem Celland Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla,CA, USA. 3Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, NIH,Bethesda, MD, USA. 4State Key Laboratory of Molecular Developmental Biology,Institute of Genetics and Developmental Biology, Chinese Academy of Sciences,Beijing, China.*Present address: Center for Cardiovascular Regeneration, Department ofCardiovascular Sciences, Houston Methodist Research Institute, Houston, TX,USA.

‡Author for correspondence ([email protected])

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

989

© 2015. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 2: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

identical to human APOC2, with 49.5% conserved consensusamino acids, but its C-terminal region (residues 67-92, zebrafishnumbering), which mediates Apoc2 binding to Lpl, shows 46.1%identity to the LPL-binding region of human APOC2. Thus, it islikely that the APOC2-LPL complex required for TG hydrolysis isalso conserved in zebrafish. To support this hypothesis, we havedeveloped an Apoc2 loss-of-function zebrafish model. Our resultsdemonstrate that the apoc2 mutant zebrafish develop severehypertriglyceridemia, which is characteristic for human patientsdeficient in APOC2, and that the apoc2 mutant is a suitable animalmodel to study hyperlipidemia and the mechanisms involved in thepathogenesis of associated diseases.

RESULTSMutation of zebrafish apoc2 gene with TALENsTo create a zebrafish model of hypertriglyceridemia, we mutated theapoc2 gene in zebrafish using a transcription activator-like effector

nuclease (TALEN) technique. We chose a target site located at exon3 of the apoc2 gene, with TALEN-binding sequences of 16 bp and17 bp nucleotides on the left and right side of the target site,respectively. The spacer DNA was 21 bp long and contained theBtsI restriction enzyme site (Fig. 1A). The corresponding proteincoding region of the TALEN target site is located in front of the Lpl-binding domain (Fig. 1B). The mRNAs encoding TALENs wereinjected into one-cell stage zebrafish embryos. To test whether theapoc2 TALEN pair disrupted the apoc2 gene at its specific targetsite, we extracted genomic DNA from F0 generation zebrafish,amplified the regions containing the target site with PCR andconducted BtsI enzyme digestion. Compared with wild-type (WT),a part of the PCR band amplified from TALEN-injected F0zebrafish was resistant to enzyme digestion (Fig. 1C), consistentwith modification of the BtsI recognition site in the apoc2 genomicsequence. Then, F0 zebrafish were outcrossed with WT zebrafishand the F1 progeny were genotyped and outcrossed again for twogenerations, followed by an incross to obtain homozygous mutants

RESOURCE IMPACT

BackgroundBlood transports dietary lipids and the lipids produced by the liver aroundthe body in the form of large lipoprotein particles, such as chylomicronsand very-low-density lipoproteins (VLDL). These lipoproteins also carryAPOC2, a protein that is needed to activate lipoprotein lipase (LPL),which hydrolyzes triglycerides to deliver fatty acids to tissues.Hypertriglyceridemia (high levels of triglycerides in the blood) is anindependent risk factor for cardiovascular disease and is positivelyassociated with metabolic disorders. Obesity, alcohol excess, lack ofexercise and an unhealthy diets can all cause hypertriglyceridemia but,in addition, patients with a genetic defect in APOC2 or LPL have severehypertriglyceridemia and often manifest skin and eye abnormalities(eruptive xanthomas and lipemia retinalis, respectively) and acute andrecurrent pancreatitis, which can be lethal. The mechanisms thatunderlie hypertriglyceridemia-associated diseases are not wellunderstood, in part, owing to the limited availability of animal models.Specifically, although substantial work has been conducted with Lpl-knockout mice, Apoc2-knockout mouse models have not been reported.

ResultsZebrafish is an emerging animal model for the study of lipid metabolismand the mechanisms of human disease related to lipid abnormalities.The advantages of using zebrafish include large progeny numbers,optical transparency of zebrafish larvae, easy genetic manipulation andcost-effective maintenance. In this study, the authors generate an apoc2mutant zebrafish characterized by the loss of Apoc2 function. Theauthors report that Apoc2 loss-of-function mutant zebrafish displaychylomicronemia (build-up of chylomicrons in the blood) and severehypertriglyceridemia, characteristics that closely resemble those seen inhuman patients with APOC2 deficiency. They show that thehypertriglyceridemia in apoc2 mutants can be rescued by injection ofplasma from wild-type zebrafish with functional Apoc2 or by injection of ahuman APOC2 mimetic peptide. Notably, apoc2 mutants fed a normaldiet accumulate lipid and lipid-laden macrophages in the vasculature,which resembles early events in the development of humanatherosclerotic lesions. Finally, apoc2 mutant embryos show ectopicovergrowth of pancreas.

Implications and future directionsTaken together, these findings indicate that the new apoc2 mutantzebrafish generated by the authors display a robust hyperlipidemiaphenotype and could, therefore, be a useful and versatile animal modelin which to study the mechanisms that underlie the human diseasesinduced by hypertriglyceridemia. Moreover, small molecule and geneticscreens using the apoc2 mutant zebrafish might suggest newapproaches to treatment of hyperlipidemia and related diseases.

Fig. 1. Generation of apoc2mutant zebrafish. (A) A diagram of partial apoc2genomic DNA sequence, showing the TALEN-binding sites (underlined) andthe spacer DNA (lowercase), which contains the BtsI restriction site.(B) A diagram of the TALEN target site in the Apoc2 protein. (C) Test forTALEN mutagenesis in F0 zebrafish with BtsI enzyme digestion. The arrowindicates themodified genomicDNA,whichwas no longer recognized and cut byBtsI. (D) Representative genotyping results of the F2 generation. (E) Sequencingdata suggest a frame shift mutation at the site preceding the LPL-binding domainin the apoc2 mutant. (F,G) apoc2mRNA expression (as assessed by qPCR) inadult liver and 5.5 dpf larvae. Results are mean±s.e.m.; n=3 in each group.**P<0.01 (Student’s t-test).

990

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 3: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

(Fig. 1D). Sequencing of the apoc2 mutant revealed a one-nucleotide replacement and ten-nucleotide deletion, resulting in aframe shift mutation changing the coding sequence of the Lpl-binding domain of the Apoc2 protein (Fig. 1E).We then isolated total RNA from the apoc2 mutant adult liver or

5.5 days post fertilization (dpf ) larvae and conducted quantitativereal-time PCR (qPCR) with primers specific to both WT andmutated apoc2 mRNA. The apoc2 mRNA was undetectable in theadult liver of the mutant (Fig. 1F) and dramatically decreased in5.5 dpf larvae (Fig. 1G). These data suggest that apoc2 mRNAtranscripts that contain the frame shift are eliminated, as has beendemonstrated for many other premature stop codon and frame shifttranscripts (Baker and Parker, 2004), and suggest that the mutationmight result in loss of Apoc2 function.

Hyperlipidemia in adult apoc2 mutantsAdult apoc2 mutants were viable and fertile, and had a normalphysical appearance (Fig. 2Aand supplementarymaterial Fig. S1). Totest whether apoc2 deficiency resulted in hypertriglyceridemia, wedrew blood from adult male zebrafish into micro-capillary tubes at 8 hafter the last feeding and let the capillaries stand overnight at 4°C tosediment erythrocytes. Plasma from an apoc2mutant showed amilkyphenotype, with a creamy layer at the top (Fig. 2B), typical of thatfound in subjectswithAPOC2orLPLdeficiency (Baggio et al., 1986;Fellin et al., 1983). These plasma samples were also subjected tonative agarose gel electrophoresis. Compared with WT, mutantplasma showed a decrease inHDL and a dramatic increase in the very-low-density lipoprotein (VLDL) and low-density lipoprotein (LDL)fraction. Chylomicrons, which were never observed in fasted WTzebrafish, were present in the mutant (Fig. 2C, gel origin). Sizeexclusion chromatography of pooled zebrafish plasma from apoc2mutants showed that there was a dramatic increase of TG and totalcholesterol (TC) in the chylomicron and VLDL fraction, as well as inLDL fraction, but decreased TC in the high-density lipoprotein (HDL)fraction, compared with that in WT zebrafish (Fig. 2D,E). Forcomparison, plasma from an Ldlr−/− mouse fed a 60% high-fat dietshowed a plasma lipoprotein profile similar to the apoc2 mutant,although it had lower TG levels. Pooled plasma from overnight fastedzebrafishwas also subjected to ultracentrifugation to float triglyceride-rich lipoproteins (TRLs). The uppermost layer, corresponding toTRLs, collected from WT and apoc2 mutant zebrafish, was thensubjected to electron microscopy to visualize lipoproteins. Althoughno TRLs were detected in the WT plasma, TRLs of different sizes,including large chylomicrons, were abundant in apoc2mutant plasma(Fig. 2F). Accordingly, plasma TG levels were dramatically increased(2488±160 mg/dl versus 67±8 mg/dl, mutant versus WT; mean±s.e.m., n=6, P<0.001, Fig. 2G) and plasma lipase activity wassignificantly decreased in apoc2 mutants compared with WT(Fig. 2H). Although it has been reported that human patients withAPOC2 deficiency have a mild elevation in plasma cholesterol(Baggio et al., 1986; Fellin et al., 1983), unexpectedly, plasma TClevelswere increased asmuchas four-fold inapoc2mutants comparedwith WT zebrafish (1202±63 mg/dl versus 296±26 mg/dl, mutantversus WT; mean±s.e.m., n=6, P<0.001) (Fig. 2I).Taken together, these data suggest that the TALEN-induced

mutation in the apoc2 gene results in severe hypertriglyceridemiaand chylomicronemia, confirming that the function of APOC2 toactivate LPL is conserved in zebrafish.

Hyperlipidemia in apoc2 mutant larvaeapoc2 mutant embryos and larvae did not show significant defectsin development, other than minor, but statistically significant,

delays in yolk utilization and growth. At 30 h post fertilization (hpf)and 3.5 dpf, the yolk in the mutant was larger but the body lengthwas shorter than that of the WT (Fig. 3A-G). However, thedevelopment stage reached at both 30 hpf and 3.5 dpf, which wasdetermined by the head position and the pigment patterns, weresimilar in mutants and WT (Fig. 3A,D).

It has been reported that transient knockdown of apoc2 inhibitsangiogenesis (Avraham-Davidi et al., 2012). To assess embryonicangiogenesis, we crossed the apoc2 mutant with fli1-EGFPtransgenic zebrafish, which express EGFP in endothelial cells. At26 hpf, there were no angiogenesis differences between apoc2mutant and WT zebrafish (Fig. 3Ha,a′). However, at 52 hpf,growth of inter-segmental blood vessels (ISV) was delayed in the

Fig. 2. Adult apoc2 mutant zebrafish develop hyperlipidemia when fed anormal diet. (A) No apparent differences between 4-month-old male WT andapoc2 mutant (mut) zebrafish. Scale bar: 0.5 cm. (B) Clear and milkyappearance of plasma from WT and apoc2 mutant zebrafish, respectively.(C) Native gel electrophoresis and neutral lipid staining of plasma fromWT andapoc2 mutant zebrafish. CM, chylomicron. (D,E) Fast protein liquidchromatography (FPLC) lipoprotein profile of pooled WT (45 animals) andapoc2 mutant (14 animals) zebrafish plasma. Plasma from an Ldlr−/− mousefed a 60% high-fat diet (HFD) was used for comparison. (F) Electronmicroscopy images of TRLs isolated from pooled WT and apoc2 mutantzebrafish plasma by ultracentrifugation as described in the Materials andMethods. (G) Plasma TG levels. Results are mean±s.e.m.; n=6 in each group;***P<0.001 (Student’s t-test). (H) Plasma lipase activity assayed as describedin the Materials and Methods. Assay buffer was used as negative control.Results aremean±s.e.m.; n=3 in each group; ***P<0.001. (I) Plasma TC levels.Results are mean±s.e.m.; n=6 in each group; ***P<0.001 (Student’s t-test).

991

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 4: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

apoc2 mutant (Fig. 3Hb,b′), as was the outgrowth of subintestinalveins (SIV) at 80 hpf (Fig. 3Hc,c′), consistent with the resultsreported by Avraham-Davidi et al. Interestingly, these ISV andSIV defects in mutants were all corrected at later stages and thevasculature of 14 dpf mutants was normal (supplementarymaterial Fig. S2).Until 5 dpf, zebrafish embryos, unlike adults, receive nutrients

through yolk utilization. To test whether TG and cholesterol levelswere affected at this early stage, which is a suitable time window fordrug screening and genetic manipulation by injection of antisensemorpholino oligonucleotides, we stained embryos at 1-6 dpf withOil Red O (ORO), which stains neutral lipids, including TG andcholesterol esters (CEs). Before 3 dpf, the strongest ORO stainingwas in the yolk and no significant differences between WT andapoc2 mutants were detected. Starting from 3 dpf, we noticedconsistently stronger vascular ORO staining in the mutants, with themost significant differences between WT and apoc2 mutantsobserved in non-fed 6 dpf embryos. At this stage, ORO staining wasweak or absent in the blood vessels of WT zebrafish, but strongORO staining was detected in the apoc2mutants (Fig. 4Aa,a′,b,b′).The dramatic differences in the levels of circulating neutral lipidswere also monitored with the fluorescent dye BODIPY (Fig. 4Ac,c′).The advantage of using BODIPY is that live zebrafish can beimmersed in the stain and after a wash, changes in neutral lipidlevels can be monitored and quantified (Fig. 4B). Furthermore, theBODIPY staining in live zebrafish persisted over a prolonged periodof time (up to 48 h in apoc2mutants in our experiments). Consistentwith the staining results, TG and TC levels were significantly

increased in the homogenates of 6 dpf apoc2 mutant larvaecompared with WT (Fig. 4C,D).

In addition to the apoc2 mutant characterized in Figs 1-4, wegenerated a second zebrafish line with a different apoc2 mutation,which introduced a stop codon in the target site, causing loss of theLpl-binding domain of Apoc2. This apoc2 mutant (line 2) alsodisplayed increased vascular ORO and BODIPY staining at 6 dpf,similar to the frame shift mutant, and the loss of apoc2 mRNA(supplementary material Fig. S3), further supporting the functionalconservation of Apoc2 in zebrafish.

Injection of normal plasma rescues hyperlipidemia in apoc2mutantsIn human patients with APOC2 deficiency, transfusions of normalplasma result in rapid and dramatic decreases in the plasma TG levelsin patients (Breckenridge et al., 1978; Nordestgaard et al., 1988). Totest whether this effect can be reproduced in our apoc2 mutantzebrafish, we isolated plasma from adult WT and mutant zebrafishand transfused them into WT and apoc2 mutant larva recipientsthrough cardinal vein injection. The plasma was supplemented withred fluorescent dextran as a tracer to confirm a successful injection.At24 h after injection, the larvaewith positive dextran fluorescencewereselected and stainedwith BODIPY. TheWT larvae injectedwithWTor mutant plasma did not show changes in BODIPY staining.Compared with apoc2 mutant plasma-injected mutant larvae, WTplasma-injected apoc2 mutant larvae showed a significant decreasein BODIPY fluorescence (Fig. 5A,B). Following BODIPY imaging,the same larvae were euthanized and homogenized, and lipids weremeasured.Consistent with the BODIPY-staining results, TG levels inthe apoc2mutants were significantly reduced following the injectionof WT plasma (Fig. 5C,D).

Fig. 3. Development and angiogenesis in apoc2 mutant larvae.(A-C) Morphology of WT (n=11) and apoc2 mutant (mut, n=11) embryosat 30 hpf. Results are mean±s.e.m.; ***P<0.001 (Student’s t-test).(D-G) Morphology of WT (n=11) and apoc2mutant (n=10) embryos at 3.5 dpf.Results are mean±s.e.m.; *P<0.05; **P<0.01; ***P<0.001 (Student’s t-test).The blue, red and yellow lengths highlighted in D were used to delineate thesize of yolk, yolk extension and the body length, respectively. (H) Angiogenesisin fli1:EGFP WT and apoc2 mutant embryos at 28 hpf (a,a′), 52 hpf (b,b′) and80 hpf (c,c′). The numbers in the figure indicate the number of animalsexhibiting the illustrated phenotype and the total number of animals. Scalebars: 200 µm (A,D); 100 µm (H).

Fig. 4. apoc2 mutant larvae develop hyperlipidemia. (A) Non-fed 6 dpf WTand apoc2 mutant (mut) larvae were stained with Oil Red O (a-b′) or BODIPY(c,c′). Scale bar: 200 µm. (B) Quantification of BODIPY staining in A. Resultsare mean±s.e.m.; n=3 in each group; ***P<0.001 (Student’s t-test).(C,D) TG and TC levels in homogenates of 6 dpf WT and apoc2mutant larvae,normalized to total protein content. Results are mean±s.e.m.; n=3 in eachgroup; **P<0.01; ***P<0.001 (Student’s t-test).

992

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 5: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

Injection of APOC2 mimetic peptide rescues hyperlipidemiain apoc2 mutantsWe have recently developed a novel bihelical amphipathic peptide(C-II-a) that contains an amphipathic helix (18A) for binding tolipoproteins and stimulating cholesterol efflux, as well as a motifbased on the last helix of humanAPOC2,which activates lipolysis byLPL (Amar et al., 2015). An inactive APOC2 peptide (C-II-i), inwhich four amino acids are mutated, was used as a negative control.The C-II-a peptide restored normal lipolysis in plasma fromAPOC2-deficient human patients and dramatically decreased the TG levels inApoe−/− mice (Amar et al., 2015). Protein alignment indicated thatnine of 20 amino acids in C-II-a were conserved between zebrafishand human, including the crucial amino acids that were mutated inC-II-i (Fig. 6A). To test whether the human APOC2-based mimeticpeptide could rescue hypertriglyceridemia in apoc2 mutantzebrafish, we injected C-II-a or C-II-i, together with a fluorescentdextran tracer, into zebrafish larvae and assessed the vascular contentof neutral lipids (using BODIPY staining) at 30 min and 6, 30 and72 h after injection. Although at 30 min there was no difference, at6 h post C-II-a injection, there was a modest, but statisticallysignificant, 20% decrease in BODIPY fluorescence (supplementarymaterial Fig. S4). Furthermore, at 30 h, the BODIPY signal wasdramatically decreased by 60% in C-II-a-injected, but not in C-II-i-injected, zebrafish (Fig. 6B,C), and the effect of the C-II-a peptidepersisted for at least 72 h (supplementary material Fig. S4).Biochemical measurements confirmed a significant decrease in TGlevels and a trend towards TC decrease (Fig. 6D,E).

Vascular lipid accumulation in apoc2 mutant larvaeIn our previous experiments, we found that supplementing a high-cholesterol diet (HCD) with red fluorescent CE enabled monitoringaccumulation of vascular lipid deposits in live transparent zebrafishlarvae (Fang et al., 2011; Stoletov et al., 2009). As apoc2 mutantzebrafish have higher TG and TC levels even when fed a diet withnormal content of cholesterol (Figs 2, 4; Fig. 7A,B), we tested theapoc2 mutants for the presence of vascular lipid deposits. WT and

apoc2 mutants were fed a diet with a normal content of fat andcholesterol from 5 to 14 dpf. To trace lipid accumulation,fluorescently labeled CE (576/589-CE; the numbers indicateexcitation/emission wavelengths) was added to the diet from 8 to12 dpf. The number of fluorescent lipid deposits was significantlyhigher in the vasculature of apoc2 mutants than in WT zebrafish(Fig. 7C,D). To test whether these vascular lipid deposits were infact intracellular lipid accumulated in macrophages, as observed inhuman and mouse early atherosclerotic lesions, we crossed apoc2mutants tompeg1-EGFP transgenic zebrafish, which express EGFPin macrophages. As expected, the majority of the lipid deposits werelocalized within macrophages (Fig. 7E,F).

Pancreatic ectopic growth in apoc2 mutantsPatients with extremely high TG levels are at greater risk for thedevelopment of acute pancreatitis (Ewald et al., 2009). However, themechanisms by which this occurs are not known with certainty,although they might be related in part to excess TG accumulation inthe pancreatic vasculature, where they would be subjected tohydrolysis by pancreatic lipases (Wang et al., 2009). To assessdevelopmental pancreatic defects, we crossed apoc2 mutants withptf1a-EGFP transgenic zebrafish, which express EGFP primarily inacinar cells (Dong et al., 2008; Godinho et al., 2005). We observedectopic outgrowth of the acinar pancreas in apoc2mutants starting at4 dpf, whichwas persistent and was also observed in 6.5 dpf embryos(Fig. 8A,B). At 6.5 dpf, a WT pancreas usually has a head domain,which contains the principal islet, and a long tail domain that extendsposteriorly. In contrast, the apoc2 mutant pancreas had additionalshort ectopic protrusions along the head of the pancreas (Fig. 8).These defects found in embryos from apoc2−/− incrosses wereconfirmed in apoc2+/− incross experiments in which embryos werescored for ectopic pancreatic outgrowth prior to genotyping (Fig. 8C).

DISCUSSIONRecent genome-wide association studies provide new strongevidence and support earlier studies suggesting that abnormal

Fig. 5. Injections of WT zebrafish plasma rescue thehyperlipidemia phenotype in apoc2mutants. (A) apoc2WT ormutant larvae at 5 dpf were injected with 5 nl of undiluted plasmafrom either WT or mutant (mut) adult male zebrafish, bothsupplemented with red fluorescent dextran. At 1 day afterinjection, at 6 dpf, larvae were stained with BODIPY (greenfluorescence) to assess the neutral lipid content of plasma. Reddextran fluorescence indicates successful injection. Scale bar:100 µm. (B) Quantification of results shown in A. Results aremean±s.e.m.; n=3 in each group; ***P<0.001; ns, not significant(Student’s t-test). (C,D) TG and TC levels in homogenates of 6 dpfWT and apoc2 mutant larvae, normalized to total protein content.Results are mean±s.e.m.; five embryos pooled per sample, n=3 ineach group; *P<0.05; **P<0.01 (Student’s t-test).

993

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 6: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

metabolism of TRLs contributes to increased risk of CVD (Do et al.,2015, 2013; Hokanson and Austin, 1996). In addition, it has beenshown that carriers of APOC3 loss-of-function mutations have 40%lower plasma TG levels and a 40% lower risk of CVD than thegeneral population (Jørgensen et al., 2014; The TG and HDLWorking Group of the Exome Sequencing Project, National Heart,Lung, and Blood Institute et al., 2014). These two Mendelianrandomization studies categorized participants based on theirAPOC3 genotype rather than plasma TG levels, theoreticallyexcluding confounding factors that affect both plasma TG levels andCVD (Cohen et al., 2014). Although one still cannot exclude thepossibility that APOC3 plays its role in CVD through otherpathways independent from its effects on TG levels, those twostudies clearly link the plasma TG levels with the genetic cause ofCVD risk.A major mechanism by which APOC3 increases plasma TG is by

its ability to inhibit LPL activity (Larsson et al., 2013), although italso is known to inhibit the clearance of VLDL and chylomicronremnants. In contrast, APOC2 is a cofactor of LPL and its loss offunction results in deficient LPL activity and consequenthypertriglyceridemia and chylomicronemia (Breckenridge et al.,1978; Cox et al., 1978). In this work, we report the development of a

new zebrafish model in which a loss-of-function mutation inthe apoc2 gene results in chylomicronemia and profoundhypertriglyceridemia. The first apoc2 mutant we selected had aframe shift mutation, which led to the amino acid sequence change inits Lpl-binding domain (Fig. 1B,E). In a different apoc2mutant line,we found a stop codon mutation, which caused deletion of the Lpl-binding domain from the Apoc2 protein sequence (supplementarymaterial Fig. S3). Importantly, both apoc2 mutant zebrafish linesshowed dramatically decreased apoc2 mRNA expression anddeveloped hypertriglyceridemia, validating the Apoc2 loss-of-function model.

The plasma of adult apoc2 mutant zebrafish shared manycharacteristics with the plasma of APOC2- or LPL-deficient humanpatients (Cox et al., 1978; Hooper et al., 2014; Okubo et al., 2014).It has significantly decreased lipase activity, chylomicronemia,dramatic hypertriglyceridemia and increased VLDL content,hypercholesterolemia and reduced levels of HDL (Fig. 2). Theadvantages of using zebrafish as an animal model include theoptical transparency of embryos and larvae, which makes themsuitable for live high-resolution imaging, and easy geneticmanipulation and drug screening. Our studies demonstrate that asearly as at 3 dpf, apoc2 mutant larvae already display remarkablehyperlipidemia, and in 6 dpf embryos, dramatic differences incirculating neutral lipids were detected with ORO staining andconfirmed with biochemical measurements of TG and TC levels inwhole-body homogenates (Fig. 4).

Fig. 7. Enhanced vascular lipid accumulation in apoc2mutants. (A,B) TGand TC levels in homogenates of 14 dpf WT and apoc2 mutant (mut) larvae,normalized to total protein content. Larvae were fed a normal diet. Results aremean±s.e.m.; n=3 in each group; *P<0.05; ***P<0.001 (Student’s t-test).(C,D) Vascular lipid deposits in 14 dpf WT (n=8) and apoc2 mutant (n=8)larvae fed a normal diet; **P<0.01 (Student’s t-test). (E) Colocalizationof vascular lipid deposits with macrophages in 14 dpf apoc2 mutantmpeg1:EGFP zebrafish. (F) Quantification of lipid deposits associated andnon-associated with macrophages in apoc2 mutant larvae. Results aremean±s.e.m.; n=7. Scale bars: 50 µm.

Fig. 6. Injections of human APOC2 mimetic peptide rescuehyperlipidemia phenotype in apoc2 mutant. (A) Protein alignment ofAPOC2mimetic peptides with the corresponding zebrafish Apoc2 amino acidssequence. Blue and red asterisks indicate the conserved amino acids in theC-II-a (active) peptide; red asterisks indicate amino acids that were mutated inthe C-II-i (inactive) peptide. (B) At 30 h after injection of 5 nl of C-II-a or C-II-i, at6.5 dpf, larvae were stained with BODIPY. Red dextran fluorescence indicatessuccessful injection. Scale bar: 50 µm. (C) Quantification of BODIPY stainingresults. Results are mean±s.e.m.; n=3 in each group; ***P<0.001; ns, notsignificant (Student’s t-test). (D,E). TG and TC levels in homogenates of6.5 dpf WT and apoc2 mutant larvae, normalized to total protein content.Results are mean±s.e.m.; five embryos pooled per sample, n=3 in each group;*P<0.05; **P<0.01; ns, not significant (Student’s t-test).

994

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 7: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

In human APOC2 patients, chylomicron and VLDL size andabundance fluctuate widely and depend on the type of foodconsumed on the preceding day (Breckenridge et al., 1978). Futurework will determine whether variation of the diet fed to apoc2mutant zebrafish will help maximize chylomicron or VLDL contentfor specific experimental goals. An important difference betweenmammalian and fish lipoprotein metabolism is the absence ofAPOBEC1 in non-mammalian vertebrates (Conticello et al., 2005;Teng and Davidson, 1992) and, thus, the absence of intestinal apoB-48, which is a major apolipoprotein in human chylomicrons. Thismight explain why the majority of TRLs we observed in apoc2mutant zebrafish were a similar size to human and mouse VLDL.The lipoprotein profiles shown in Fig. 2D-F suggest that the apoc2mutant zebrafish are suitable for modeling human dyslipidemia, andthe zebrafish model favorably compares with a large number ofother models, including the Ldlr−/− mouse, as profiled previously(Yin et al., 2012).In this work, we developed a simple and convenient protocol for

staining circulating lipoproteins with BODIPY. BODIPY is alipophilic fluorescent dye, which preferentially localizes to theneutral lipid core of lipoproteins and intracellular lipid droplets, andis widely used in cell biology (Bozaquel-Morais et al., 2010; Goczeand Freeman, 1994; Grandl and Schmitz, 2010). However, to thebest of our knowledge, BODIPY has not been used to stain andmonitor circulating lipid levels in live zebrafish larvae. In ourexperiments, live zebrafish were immersed in a BODIPY solution,with a subsequent wash in water. This simple procedure resulted inbright green fluorescent staining of neutral lipids in the circulationof apoc2 mutant animals. This assay provides a robust readout andmight help conduct genetic and drug screening studies.Weused theBODIPYassay to demonstrate that injections ofnormal

zebrafish plasma significantly diminished hypertriglyceridemia inapoc2mutant zebrafish (Fig. 5). This result is similar to the treatmentof APOC2-deficient patients with severe hypertriglyceridemia inwhich transfusions of normal, APOC2-containing plasma leads torapid (within 1 day) reductions in plasma TGs (Breckenridge et al.,1978). Remarkably, the amino acids that are crucial for LPLactivation in the last helix of human APOC2 are conserved inzebrafish, and the human APOC2 mimetic peptide C-II-a (Amaret al., 2015), rescued severe hypertriglyceridemia in apoc2 mutantzebrafish (Fig. 6). These results suggest that our zebrafish model issuitable for testing potential therapeutic agents for treatment ofhuman APOC2 deficiency.

Two previous studies in which apoc2 was transiently knockeddown in zebrafish embryos with antisense morpholinooligonucleotides, have suggested that Apoc2 is involved in yolkabsorption and angiogenesis (Avraham-Davidi et al., 2012; Pickartet al., 2006). Our results with the apoc2 mutant zebrafish supportthese conclusions. Consistent with the results reported by Pickartet al., apoc2mutant larvae show a slightly delayed yolk consumption(Fig. 3A-G) and the mutant larvae are smaller than WT larvae. Thisphenotype in the apoc2 mutant can be explained, in part, by thedisruption of the Lpl function, resulting in a defect in TG hydrolysisand the supplyof free fatty acids (FFAs) to non-hepatic tissues,which,in turn, might cause nutrient deprivation and delayed growth. Incontrast, by monitoring the head angle at 28 hpf and the pattern ofpigmentation at 3 dpf, we did not notice any differences in the stage ofdevelopment reached betweenWTand apoc2mutants. Because adultapoc2mutant zebrafish are the same size asWTand have no apparentphenotype (Fig. 2A; supplementary material Fig. S1), other thanhyperlipidemia, it is possible that feeding and the activity of otherlipases help restore the energy balance in unchallenged animals.

A previous study has reported that a transient apoc2 knockdownby antisense morpholino oligonucleotides increases levels of apoB-containing lipoproteins, which, in turn, inhibits embryonicangiogenesis (Avraham-Davidi et al., 2012). In our experiments,the Apoc2 loss of function resulted in dramatically increased levelsof VLDL, an apoB-containing lipoprotein, and indeed delayed ISVand SIV growth. Nevertheless, the apoc2 mutants were viable andtheir vascular structure was normal at 14 dpf despite profoundhyperlipidemia (supplementary material Fig. S2). Thus, the excessof apoB-containing lipoproteins is likely to delay but not tocompletely inhibit angiogenesis in vivo.

In agreement with epidemiologic studies suggesting thathypertriglyceridemia is a risk factor for CVD (Do et al., 2015;Jørgensen et al., 2014;TheTGandHDLWorkingGroupof theExomeSequencing Project, National Heart, Lung, and Blood Instituteet al., 2014), our results demonstrate that the hypertriglyceridemiainduced by the apoc2 mutation in zebrafish results in vascular lipidaccumulation and macrophage lipid uptake (Fig. 7C-E), whichare important initial events in the development of humanatherosclerosis. The vascular lipid accumulation in zebrafishinduced by hypertriglyceridemia was similar to that inducedby hypercholesterolemia (O’Hare et al., 2014; Stoletov et al.,2009). Our study is also consistent with mouse studies in whichhypertriglyceridemia induced by Lpl orGpihbp knockout resulted in

Fig. 8. apoc2 mutants have ectopic pancreas outgrowth. (A) Images of pancreas, labeled with EGFP driven by the ptf1a promoter, in 6.5 dpf WT and apoc2mutant (mut) larvae. The yellow arrow points to the normal pancreatic growth and white arrows point to ectopic outgrowth. Scale bar: 100 µm. (B,C) At 6.5 dpf, anindependent observer scored a pancreatic outgrowth phenotype as 0, normal, 1, mild and 2, severe. (B) WT (n=13) and apoc2−/− mutant (n=21). Results aremean±s.e.m.; ***P<0.001 (Student’s t-test). (C) Heterozygous apoc2+/− zebrafish carrying one copy of the ptf1a-EGFP transgene were crossed withthe heterozygous apoc2+/− zebrafish carrying no ptf1a-EGFP. A total of 22 embryos that displayed EGFP fluorescence were selected, scored for thepancreas outgrowth defect and then genotyped. Random ptf1a-EGFP expression resulted in the following distribution of EGFP-positive embryos: apoc2+/+ (n=3),apoc2+/− (n=9) and apoc2−/− (n=10). Results are mean±s.e.m.; *P<0.05; ***P<0.001 (Student’s t-test).

995

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 8: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

spontaneous development of atherosclerosis (Weinstein et al., 2010;Zhang et al., 2008). In contrast to hypertriglyceridemic mice, inwhich atherosclerotic lesions develop late, by the age of 1 year,vascular lipid deposits in apoc2 mutant zebrafish fed normal dietdeveloped as early as at 14 dpf. One possible explanation is theabsence of CETP inmice, whereas zebrafish express functionalCetp(Jin et al., 2011; Kim et al., 2012). Transfer of cholesterol fromHDLto LDL and VLDL (and TG in the opposite direction) might play anatherogenic role in apoc2 mutant zebrafish, as it does in humans(Barter and Rye, 2012).One important clinical complication of hypertriglyceridemia in

patients with LPL or APOC2 deficiency is recurrent pancreatitis, butthe underlying mechanisms are poorly defined (Baggio et al., 1986;Fellin et al., 1983). Inapoc2mutant zebrafish,we found ectopic acinaroutgrowth in the head of the pancreas (Fig. 8). Acinar cells expresshigh levels of pancreatic lipase. Chylomicronemia results in increasedFFA uptake and injury in the acinar cells, which might lead toinflammation and pancreatitis (Scherer et al., 2014; Wang et al.,2009). Our findings suggest that the Apoc2 deficiency affects earlypancreas development in zebrafish. A recent report shows thatmodeling chylomicron retention disease in zebrafish,whichmanifestsin the reduction of circulating neutral lipids, was associated withinhibited growth of exocrine pancreas (Levic et al., 2015). We as yetdo not know whether the hyper- or hypo-triglyceridemia effects onexocrine pancreas growth in apoc2mutant zebrafish are related to thepathogenesis of pancreatitis in human APOC2-deficient patients.In summary, our new apoc2 mutant zebrafish display a robust

hyperlipidemia phenotype and present a useful and versatile animalmodel to study mechanisms related to human diseases induced byhypertriglyceridemia. Small-molecule and genetic screens using theapoc2 mutant zebrafish might suggest new approaches to treatmentof hyperlipidemia and related disorders.

MATERIALS AND METHODSZebrafish maintenance and feedingAdult zebrafish of the AB strain were maintained at 28°C on a 14-h-light–10-h-dark cycle and fed brine shrimp twice a day. Zebrafish larvae were fedGolden Pearls (100–200 µm size from Brine Shrimp Direct, Ogden, UT)twice a day, starting from 4.5 dpf. For lipid-deposition experiments, GoldenPearls, supplemented with 1 µg/g of a fluorescent cholesterol ester analog(cholesteryl BODIPY 576/589-C11 from Invitrogen, Carlsbad, CA; catalognumber C12681), were fed to zebrafish from 8 to 12 dpf as describedpreviously (Stoletov et al., 2009). fli1:EGFP and mpeg1-EGFP transgeniczebrafish were from the Weinstein (Lawson and Weinstein, 2002) andLieschke (Ellett et al., 2011) laboratories, respectively. ptf1a-EGFPtransgenic zebrafish were developed in the Dong laboratory (Dong et al.,2008; Godinho et al., 2005). All animal studies were approved by the UCSDInstitutional Animal Care and Use Committee.

TALEN construction, mRNA synthesis, injection andconfirmation of apoc2 mutationTALEN plasmids targeting apoc2 were constructed from the stock cassetteaccording to the published protocols (Dong et al., 2011; Huang et al., 2014).TALEN mRNAs were synthesized with an mMESSAGE mMACHINE Sp6transcription kit (Ambion, Austin, TX; AM1340). mRNAs encoding thepair of TALEN proteins were mixed at a 1:1 ratio to a final concentration of200 ng/µl. A total of 1-2 nl of TALENmRNAswas injected into the one-cellstage embryos. Genomic DNA (gDNA) was extracted from whole embryosor from the tissue clipped off the tail fin of adult zebrafish and wasgenotyped by PCR (with the following primers: forward, 5′-GTGGTCG-CATAGTTTAAGCT-3′; reverse, 5′-GCAACCATGTAAGAGTTGCA-3′)and BtsI enzyme digestion, and confirmed by sequencing, according topublished protocols (Dong et al., 2011; Huang et al., 2014). To assessexpression of apoc2 in larvae homogenates and in adult liver, total RNAwas

isolated, reverse transcribed and subjected to qPCR with the followingprimers: forward, 5′-ATGAACAAGATACTGGCTAT-3′; reverse, 5′-TTGATGGTCTCTACATATCC-3′, using a KAPA SYBR FASTUniversal qPCR kit (KAPA Biosystems, Wilmington, MA; KK4602) anda Rotor Gene Q qPCR machine (Qiagen, Valencia, CA). The position ofthese qPCR primers relative to the TALEN target site is shown insupplementary material Fig. S3. Because the mutation does not changenucleotide sequence beyond the target site, these primers detect expressionof both WT and mutant mRNA, if any is present.

Oil red O staining and BODIPY stainingOil red O (ORO) staining was conducted according to published protocols(Schlegel and Stainier, 2006). Briefly, embryos were fixed in 4%paraformaldehyde (PFA) for 2 h, washed three times in PBS, incubated in0.3% ORO solution for 2 h and then washed with PBS before imaging. ForBODIPY staining, live larvae were immersed in E3 water (5.0 mM NaCl,0.17 mM KCl, 0.33 mM CaCl2, 0.33 mM MgSO4) containing 0.1 µg/mlBODIPY® 505/515 (4,4-difluoro-1,3,5,7-tetramethyl-4-Bora-3a,4a-diaza-s-indacene, Invitrogen; D-3921) for 1 h in dark and then rinsed with E3water before imaging.

Triglyceride and cholesterol measurements and lipoproteinanalysisBlood was collected from adult male zebrafish, after overnight fasting,through tail amputation and diluted 1:50 (WT) or 1:200 (apoc2 mutant) inPBS.The supernatantswere collected as a plasmafraction after centrifugationat 2350 g for 10 min. Embryos or larvae were gently homogenized in PBSwith a pestle. After centrifugation at 16,000 g for 10 min, supernatants werecollected and referred to as ‘homogenates’. TG and TC levels were measuredin 25 µl of diluted plasma or embryo or larva homogenates using kits fromBioVision (Milpitas, CA; Triglyceride Quantification Kit, K622-100;Cholesterol Quantification Kit, K623-100) and according to themanufacturer’s protocol. To assess undiluted plasma, 5 µl blood wascollected in a tube containing 0.5 µl heparin (5 mg/ml) and then loadedinto a heparin-rinsed capillary, which was kept overnight at 4°C in a verticalposition. Following erythrocyte sedimentation, the capillaries werephotographed. Lipoprotein fractions were assessed using native agarose gelelectrophoresis (Helena Laboratories, Beaumont, TX; 3045) as wepreviously described (Stoletov et al., 2009).

Fast protein liquid chromatography lipoprotein profileA total of 100 µl of pooled plasma from 45 adult WT zebrafish or 40 µl ofpooled plasma from 14 adult apoc2 mutant zebrafish were loaded onto aSuperose 6 PC 3.2/30 column (GE Healthcare Life Science, Pittsburgh, PA;17-0673-01), and TC and TG levels were determined in each fraction(250 μl), collected at a flow speed of 0.5 ml/min.

Ultracentrifugation and electron microscopyPooled plasma from fasted overnight WT and apoc2 mutant zebrafish wasdiluted in PBS, without density adjustment, and the samples werecentrifuged at 200,000 g for 4 h at 4°C. The top layer containing TRLswas collected, supplemented with 0.1% sucrose, added to a FCF100-Cu 100mesh (Electron Microscopy Sciences, Hatfield, PA) and negatively stainedwith 1% uranyl acetate. Stained samples were imaged with a Tecnai G2Spirit BioTWIN transmission electron microscope equipped with a 4KEagle digital camera (FEI Company, Hillsboro, OR).

Plasma lipase activity assayAdult zebrafish blood was diluted 1:25 in PBS and after centrifugation, 5 µlof the diluted plasmawas used to measure lipase assay with a Lipase activityassay kit (Cayman Chemical, Ann Arbor, MI; 700640), following themanufacturer’s manual. The assay buffer was used as a negative control. Thereaction was conducted at 30°C.

Intravenous injectionsBlood was collected by tail amputation from adult male zebrafish at 4 h afterthe last feeding, and plasma was separated by centrifugation at 2350 g for

996

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 9: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

10 min. C-II-a and C-II-i peptides (Amar et al., 2015) were dissolved in PBS(pH 7.4) to a concentration of 2 mg/ml. To ensure visual control ofsuccessful injection, zebrafish plasma or APOC2 mimetic peptides weresupplemented with 2% (v/v) tetramethylrhodamine-conjugated 10 kDadextran (0.5 mg/ml miniRuby, Invitrogen, D-3312). For injections, 5 dpflarvae were aligned in 0.5% low-melting-point agarose. A total of 5 nl ofplasma or APOC2 mimetic peptides were injected through the cardinal veinabove the heart chamber using a FemtoJet microinjector (Eppendorf,Hamburg, Germany). The red signal from fluorescent dextran confirmedsuccessful injection.

Imaging of live embryos or larvaeFor in vivo live microscopy, anesthetized embryos or larvaewere mounted inlow-melting-point agarose (0.5%, Fisher, Pittsburgh, PA; BP1360-100)containing tricaine (0.02%, Sigma, St Louis, MO; A5040) in 50-mm glass-bottom dishes (MatTek, Ashland, MA; P50G-0-14-F). Images werecaptured with Leica CTR5000 (Wetzlar, Germany), Olympus FV1000spectral confocal (Tokyo, Japan) or BZ9000 Keyence (Osaka, Japan)fluorescent microscopes.

Competing interestsJ.L.W. is a consultant for ISIS Pharmaceuticals. Other authors declare no competinginterests.

Author contributionsC.L. and Y.I.M conceived and designed the experiments. C.L., K.P.G., L.F., D.A.S.,H.G., J.P.,W.H. and J.K. performed the experiments. M.J.A., J.Z., J.L.W., A.T.R. andP.D.S.D. provided crucial materials, ideas and discussion for this work. C.L. andY.I.M. wrote the paper.

FundingThe project was supported by the National Institutes of Health (NIH) [grant numbersHL093767 to Y.I.M., HL055798 to Y.I.M., HL088093 to J.L.W. and DK098092P.D.S.D.], as well as the UCSD Neuroscience Microscopy Facility [grant numberP30NS0471010].

Supplementary materialSupplementary material available online athttp://dmm.biologists.org/lookup/suppl/doi:10.1242/dmm.019836/-/DC1

ReferencesAmar,M. J. A., Sakurai, T., Sakurai-Ikuta, A., Sviridov, D., Freeman, L., Ahsan, L.and Remaley, A. T. (2015). A novel apolipoprotein C-II mimetic peptide thatactivates lipoprotein lipase and decreases serum triglycerides in apolipoprotein E-knockout mice. J. Pharmacol. Exp. Ther. 352, 227-235.

Anderson, J. L., Carten, J. D. and Farber, S. A. (2011). Zebrafish lipid metabolism:from mediating early patterning to the metabolism of dietary fat and cholesterol.Methods Cell Biol. 101, 111-141.

Avraham-Davidi, I., Ely, Y., Pham, V. N., Castranova, D., Grunspan, M.,Malkinson, G., Gibbs-Bar, L., Mayseless, O., Allmog, G., Lo, B. et al.(2012). ApoB-containing lipoproteins regulate angiogenesis by modulatingexpression of VEGF receptor 1. Nat. Med. 18, 967-973.

Baggio, G., Manzato, E., Gabelli, C., Fellin, R., Martini, S., Enzi, G. B., Verlato, F.,Baiocchi, M. R., Sprecher, D. L., Kashyap, M. L. et al. (1986). Apolipoprotein C-II deficiency syndrome. Clinical features, lipoprotein characterization, lipaseactivity, and correction of hypertriglyceridemia after apolipoprotein C-IIadministration in two affected patients. J. Clin. Invest. 77, 520-527.

Baker, K. E. and Parker, R. (2004). Nonsense-mediated mRNA decay: terminatingerroneous gene expression. Curr. Opin. Cell Biol. 16, 293-299.

Barter, P. J. and Rye, K. A. (2012). Cholesteryl ester transfer protein inhibition as astrategy to reduce cardiovascular risk. J. Lipid Res. 53, 1755-1766.

Bozaquel-Morais, B. L., Madeira, J. B., Maya-Monteiro, C. M., Masuda, C. A. andMontero-Lomeli, M. (2010). A new fluorescence-based method identifies proteinphosphatases regulating lipid droplet metabolism. PLoS ONE 5, e13692.

Breckenridge, W. C., Little, J. A., Steiner, G., Chow, A. and Poapst, M. (1978).Hypertriglyceridemia associated with deficiency of apolipoprotein C-II.N. Engl. J. Med. 298, 1265-1273.

Cohen, J. C., Stender, S. and Hobbs, H. H. (2014). APOC3, coronary disease, andcomplexities of Mendelian Randomization. Cell Metab. 20, 387-389.

Conticello, S. G., Thomas, C. J., Petersen-Mahrt, S. K. and Neuberger, M. S.(2005). Evolution of the AID/APOBEC family of polynucleotide (deoxy)cytidinedeaminases. Mol. Biol. Evol. 22, 367-377.

Cox, D. W., Breckenridge, W. C. and Little, J. A. (1978). Inheritance ofapolipoprotein C-II deficiency with hypertriglyceridemia and pancreatitis.N. Engl. J. Med. 299, 1421-1424.

Do, R., Willer, C. J., Schmidt, E. M., Sengupta, S., Gao, C., Peloso, G. M.,Gustafsson, S., Kanoni, S., Ganna, A., Chen, J. et al. (2013). Common variantsassociated with plasma triglycerides and risk for coronary artery disease. Nat.Genet. 45, 1345-1352.

Do, R., Stitziel, N. O., Won, H.-H., Jørgensen, A. B., Duga, S., Angelica Merlini,P., Kiezun, A., Farrall, M., Goel, A., Zuk, O. et al. (2015). Exome sequencingidentifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction.Nature 518, 102-106.

Dong, P. D. S., Provost, E., Leach, S. D. and Stainier, D. Y. R. (2008). Gradedlevels of Ptf1a differentially regulate endocrine and exocrine fates in thedeveloping pancreas. Genes Dev. 22, 1445-1450.

Dong, Z., Ge, J., Li, K., Xu, Z., Liang, D., Li, J., Li, J., Jia, W., Li, Y., Dong, X. et al.(2011). Heritable targeted inactivation of myostatin gene in yellow catfish(Pelteobagrus fulvidraco) using engineered zinc finger nucleases. PLoS ONE 6,e28897.

Dooley, K. and Zon, L. I. (2000). Zebrafish: a model system for the study of humandisease. Curr. Opin. Genet. Dev. 10, 252-256.

Ebara, T., Ramakrishnan, R., Steiner, G. and Shachter, N. S. (1997).Chylomicronemia due to apolipoprotein CIII overexpression in apolipoproteinE-null mice. Apolipoprotein CIII-induced hypertriglyceridemia is not mediatedby effects on apolipoprotein E. J. Clin. Invest. 99, 2672-2681.

Ellett, F., Pase, L., Hayman, J. W., Andrianopoulos, A. and Lieschke, G. J.(2011). mpeg1 promoter transgenes direct macrophage-lineage expression inzebrafish. Blood 117, e49-e56.

Ewald, N., Hardt, P. D. and Kloer, H.-U. (2009). Severe hypertriglyceridemia andpancreatitis: presentation and management. Curr. Opin. Lipidol. 20, 497-504.

Fang, L., Green, S. R., Baek, J. S., Lee, S.-H., Ellett, F., Deer, E., Lieschke, G. J.,Witztum, J. L., Tsimikas, S. and Miller, Y. I. (2011). In vivo visualization andattenuation of oxidized lipid accumulation in hypercholesterolemic zebrafish.J. Clin. Invest. 121, 4861-4869.

Fang, L., Liu, C. and Miller, Y. I. (2014). Zebrafish models of dyslipidemia:relevance to atherosclerosis and angiogenesis. Transl. Res. 163, 99-108.

Fellin, R., Baggio, G., Poli, A., Augustin, J., Baiocchi, M. R., Baldo, G.,Sinigaglia, M., Greten, H. and Crepaldi, G. (1983). Familial lipoprotein lipaseand apolipoprotein C-II deficiency Lipoprotein and apoprotein analysis, adiposetissue and hepatic lipoprotein lipase levels in seven patients and their first degreerelatives. Atherosclerosis 49, 55-68.

Fukushima, N. and Yamamoto, K. (2004). [Apolipoprotein C-I, C-II, C-III]. NihonRinsho 62 Suppl. 12, 110-113.

Garcia-Arcos, I., Hiyama, Y., Drosatos, K., Bharadwaj, K. G., Hu, Y., Son, N. H.,O’Byrne, S. M., Chang, C. L., Deckelbaum, R. J., Takahashi, M. et al. (2013).Adipose-specific lipoprotein lipase deficiency more profoundly affects brown thanwhite fat biology. J. Biol. Chem. 288, 14046-14058.

Gocze, P. M. and Freeman, D. A. (1994). Factors underlying the variability of lipiddroplet fluorescence in MA-10 Leydig tumor cells. Cytometry 17, 151-158.

Godinho, L., Mumm, J. S., Williams, P. R., Schroeter, E. H., Koerber, A., Park,S.W., Leach, S. D. andWong, R. O. L. (2005). Targeting of amacrine cell neuritesto appropriate synaptic laminae in the developing zebrafish retina. Development132, 5069-5079.

Goldberg, I. J. andMerkel, M. (2001). Lipoprotein lipase: physiology, biochemistry,and molecular biology. Front. Biosci. 6, d388-d405.

Goldberg, I. J., Eckel, R. H. and McPherson, R. (2011). Triglycerides and heartdisease: still a hypothesis? Arterioscler. Thromb. Vasc. Biol. 31, 1716-1725.

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. (2014). TheGPIHBP1-LPL complex is responsible for the margination of triglyceride-richlipoproteins in capillaries. Cell Metab. 19, 849-860.

Grandl, M. and Schmitz, G. (2010). Fluorescent high-content imaging allows thediscrimination and quantitation of E-LDL-induced lipid droplets and Ox-LDL-generated phospholipidosis in human macrophages. Cytometry A 77, 231-242.

Hokanson, J. E. and Austin, M. A. (1996). Plasma triglyceride level is a risk factorfor cardiovascular disease independent of high-density lipoprotein cholesterollevel: a meta-analysis of population-based prospective studies. J. Cardiovasc.Risk 3, 213-219.

Holtta-Vuori, M., Salo, V. T. V., Nyberg, L., Brackmann, C., Enejder, A., Panula, P.and Ikonen, E. (2010). Zebrafish: gaining popularity in lipid research.Biochem. J.429, 235-242.

Hooper, A. J., Kurtkoti, J., Hamilton-Craig, I. and Burnett, J. R. (2014). Clinicalfeatures and genetic analysis of three patients with severe hypertriglyceridaemia.Ann. Clin. Biochem. 51, 485-489.

Huang, P., Xiao, A., Tong, X., Zu, Y., Wang, Z. and Zhang, B. (2014). TALENconstruction via “Unit Assembly” method and targeted genome modifications inzebrafish. Methods 69, 67-75.

Jin, S., Hong, J.-H., Jung, S.-H. and Cho, K.-H. (2011). Turmeric and laurelaqueous extracts exhibit in vitro anti-atherosclerotic activity and in vivohypolipidemic effects in a zebrafish model. J. Med. Food 14, 247-256.

997

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms

Page 10: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · hypertriglyceridemia while consuming a normal diet, often manifesting xanthomas, lipemia retinalis and pancreatitis

Jørgensen, A. B., Frikke-Schmidt, R., Nordestgaard, B. G. and Tybjaerg-Hansen, A. (2014). Loss-of-function mutations in APOC3 and risk of ischemicvascular disease. N. Engl. J. Med. 371, 32-41.

Kei, A. A., Filippatos, T. D., Tsimihodimos, V. and Elisaf, M. S. (2012). A review ofthe role of apolipoprotein C-II in lipoprotein metabolism and cardiovasculardisease. Metabolism 61, 906-921.

Kim, J. Y., Hong, J. H., Jung, H. K., Jeong, Y. S. and Cho, K. H. (2012). Grape skinand loquat leaf extracts and acai puree have potent anti-atherosclerotic and anti-diabetic activity in vitro and in vivo in hypercholesterolemic zebrafish. Int. J. Mol.Med. 30, 606-614.

Larsson, M., Vorrsjo, E., Talmud, P., Lookene, A. and Olivecrona, G. (2013).Apolipoproteins C-I and C-III inhibit lipoprotein lipase activity by displacement ofthe enzyme from lipid droplets. J. Biol. Chem. 288, 33997-34008.

Lawson, N. D. and Weinstein, B. M. (2002). In vivo imaging of embryonic vasculardevelopment using transgenic zebrafish. Dev. Biol. 248, 307-318.

Levak-Frank, S., Hofmann, W., Weinstock, P. H., Radner, H., Sattler, W.,Breslow, J. L. and Zechner, R. (1999). Induced mutant mouse lines that expresslipoprotein lipase in cardiac muscle, but not in skeletal muscle and adipose tissue,have normal plasma triglyceride and high-density lipoprotein-cholesterol levels.Proc. Natl. Acad. Sci. USA 96, 3165-3170.

Levic, D. S., Minkel, J. R., Wang, W.-D., Rybski, W. M., Melville, D. B. andKnapik, E. W. (2015). Animal model of Sar1b deficiency presents lipid absorptiondeficits similar to Anderson disease. J. Mol. Med. 93, 165-176.

Merkel, M., Weinstock, P. H., Chajek-Shaul, T., Radner, H., Yin, B., Breslow, J. L.and Goldberg, I. J. (1998). Lipoprotein lipase expression exclusively in liver. Amouse model for metabolism in the neonatal period and during cachexia. J. Clin.Invest. 102, 893-901.

Miller, M., Stone, N. J., Ballantyne, C., Bittner, V., Criqui, M. H., Ginsberg, H. N.,Goldberg, A. C., Howard, W. J., Jacobson, M. S., Kris-Etherton, P. M. et al.(2011). Triglycerides and cardiovascular disease: a scientific statement from theAmerican Heart Association. Circulation 123, 2292-2333.

Namboodiri, K. K., Kaplan, E. B., Heuch, I., Elston, R. C., Green, P. P., Rao, D. C.,Laskarzewski, P., Glueck, C. J. and Rifkind, B. M. (1985). The CollaborativeLipid Research Clinics Family Study: biological and cultural determinants offamilial resemblance for plasma lipids and lipoproteins. Genet. Epidemiol. 2,227-254.

Nordestgaard, B. G., Stender, S. and Kjeldsen, K. (1988). Reducedatherogenesis in cholesterol-fed diabetic rabbits. Giant lipoproteins do not enterthe arterial wall. Arterioscler. Thromb. Vasc. Biol. 8, 421-428.

O’Hare, E. A., Wang, X., Montasser, M. E., Chang, Y.-P. C., Mitchell, B. D. andZaghloul, N. A. (2014). Disruption of ldlr causes increased LDL-c and vascularlipid accumulation in a zebrafish model of hypercholesterolemia. J. Lipid Res. 55,2242-2253.

Okubo, M., Toromanovic, A., Ebara, T. andMurase, T. (2014). Apolipoprotein C-II:a novel large deletion in APOC2 caused by Alu-Alu homologous recombination inan infant with apolipoprotein C-II deficiency. Clin. Chim. Acta 438C, 148-153.

Otis, J. P., Zeituni, E. M., Thierer, J. H., Anderson, J. L., Brown, A. C., Boehm,E. D., Cerchione, D. M., Ceasrine, A. M., Avraham-Davidi, I., Tempelhof, H.et al. (2015). Zebrafish as a model for apolipoprotein biology: comprehensiveexpression analysis and a role for ApoA-IV in regulating food intake. Dis. Model.Mech. 8, 295-309.

Pickart, M. A., Klee, E. W., Nielsen, A. L., Sivasubbu, S., Mendenhall, E. M., Bill,B. R., Chen, E., Eckfeldt, C. E., Knowlton, M., Robu, M. E. et al. (2006).

Genome-wide reverse genetics framework to identify novel functions of thevertebrate secretome. PLoS ONE 1, e104.

Scherer, J., Singh, V. P., Pitchumoni, C. S. and Yadav, D. (2014). Issues inhypertriglyceridemic pancreatitis: an update. J. Clin. Gastroenterol. 48, 195-203.

Schlegel, A. and Stainier, D. Y. R. (2006). Microsomal triglyceride transfer proteinis required for yolk lipid utilization and absorption of dietary lipids in zebrafishlarvae. Biochemistry 45, 15179-15187.

Shachter, N. S., Hayek, T., Leff, T., Smith, J. D., Rosenberg, D. W., Walsh, A.,Ramakrishnan, R., Goldberg, I. J., Ginsberg, H. N. and Breslow, J. L. (1994).Overexpression of apolipoprotein CII causes hypertriglyceridemia in transgenicmice. J. Clin. Invest. 93, 1683-1690.

Stoletov, K., Fang, L., Choi, S.-H., Hartvigsen, K., Hansen, L. F., Hall, C.,Pattison, J., Juliano, J., Miller, E. R., Almazan, F. et al. (2009). Vascular lipidaccumulation, lipoprotein oxidation, and macrophage lipid uptake inhypercholesterolemic zebrafish. Circ. Res. 104, 952-960.

Surendran, R. P., Visser, M. E., Heemelaar, S., Wang, J., Peter, J., Defesche,J. C., Kuivenhoven, J. A., Hosseini, M., Peterfy, M., Kastelein, J. J. P. et al.(2012). Mutations in LPL, APOC2, APOA5, GPIHBP1 and LMF1 in patients withsevere hypertriglyceridaemia. J. Intern. Med. 272, 185-196.

Teng, B. andDavidson, N. O. (1992). Evolution of intestinal apolipoprotein BmRNAediting. Chicken apolipoprotein B mRNA is not edited, but chicken enterocytescontain in vitro editing enhancement factor(s). J. Biol. Chem. 267, 21265-21272.

The TG and HDL Working Group of the Exome Sequencing Project, NationalHeart, Lung, and Blood Institute, Crosby, J., Peloso, G. M., Auer, P. L.,Crosslin, D. R., Stitziel, N. O., Lange, L. A., Lu, Y., Tang, Z. Z., Zhang, H.,Hindy, G. et al. (2014). Loss-of-function mutations in APOC3, triglycerides, andcoronary disease. N. Engl. J. Med. 371, 22-31.

Trent, C. M., Yu, S., Hu, Y., Skoller, N., Huggins, L. A., Homma, S. andGoldberg,I. J. (2014). Lipoprotein lipase activity is required for cardiac lipid dropletproduction. J. Lipid Res. 55, 645-658.

Wang, Y., Sternfeld, L., Yang, F., Rodriguez, J. A., Ross, C., Hayden, M. R.,Carriere, F., Liu, G., Hofer, W. and Schulz, I. (2009). Enhanced susceptibility topancreatitis in severe hypertriglyceridaemic lipoprotein lipase-deficient mice andagonist-like function of pancreatic lipase in pancreatic cells. Gut 58, 422-430.

Watts, G. F., Ooi, E. M. M. and Chan, D. C. (2013). Demystifying the managementof hypertriglyceridaemia. Nat. Rev. Cardiol. 10, 648-661.

Weinstein, M. M., Yin, L., Tu, Y., Wang, X., Wu, X., Castellani, L. W., Walzem,R. L., Lusis, A. J., Fong, L. G., Beigneux, A. P. et al. (2010). Chylomicronemiaelicits atherosclerosis in mice–brief report. Arterioscler. Thromb. Vasc. Biol. 30,20-23.

Weinstock, P. H., Bisgaier, C. L., Aalto-Setala, K., Radner, H., Ramakrishnan, R.,Levak-Frank, S., Essenburg, A. D., Zechner, R. and Breslow, J. L. (1995).Severe hypertriglyceridemia, reduced high density lipoprotein, and neonatal deathin lipoprotein lipase knockout mice. Mild hypertriglyceridemia with impaired verylow density lipoprotein clearance in heterozygotes. J. Clin. Invest. 96, 2555-2568.

Yin, W., Carballo-Jane, E., McLaren, D. G., Mendoza, V. H., Gagen, K.,Geoghagen, N. S., McNamara, L. A., Gorski, J. N., Eiermann, G. J., Petrov, A.et al. (2012). Plasma lipid profiling across species for the identification of optimalanimal models of human dyslipidemia. J. Lipid Res. 53, 51-65.

Zhang, X., Qi, R., Xian, X., Yang, F., Blackstein, M., Deng, X., Fan, J., Ross, C.,Karasinska, J., Hayden, M. R. et al. (2008). Spontaneous atherosclerosis inaged lipoprotein lipase-deficient mice with severe hypertriglyceridemia on anormal chow diet. Circ. Res. 102, 250-256.

998

RESOURCE ARTICLE Disease Models & Mechanisms (2015) 8, 989-998 doi:10.1242/dmm.019836

Disea

seModels&Mechan

isms