apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human lpl in either...

26
© 2015. Published by The Company of Biologists Ltd. 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. 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 Si Dong 2 , Yury I. Miller 1* 1 Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA 2 Sanford Children’s 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 # Current address: Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA * To whom correspondence should be addressed at: 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 858-822-5771. Fax: 858-534-2005. Email: [email protected] Keywords: zebrafish, apolipoprotein C-II, lipoprotein lipase, hyperlipidemia Disease Models & Mechanisms DMM Accepted manuscript http://dmm.biologists.org/lookup/doi/10.1242/dmm.019836 Access the most recent version at DMM Advance Online Articles. Posted 4 June 2015 as doi: 10.1242/dmm.019836 http://dmm.biologists.org/lookup/doi/10.1242/dmm.019836 Access the most recent version at First posted online on 4 June 2015 as 10.1242/dmm.019836

Upload: others

Post on 30-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

© 2015. Published by The Company of Biologists Ltd.

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.

Apoc2 loss-of-function zebrafish mutant as a genetic model of

hyperlipidemia

Chao Liu1, Keith P. Gates2, Longhou Fang1#, Marcelo J. Amar3, Dina A. Schneider1,

Honglian Geng1, Wei Huang1, Jungsu Kim1, Jennifer Pattison1, Jian Zhang4, Joseph L.

Witztum1, Alan T. Remaley3, P. Duc Si Dong2, Yury I. Miller1*

1 Division of Endocrinology and Metabolism, Department of Medicine, University of

California, San Diego, La Jolla, CA, USA 2 Sanford Children’s 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

# Current address: Center for Cardiovascular Regeneration, Department of

Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA

* To whom correspondence should be addressed at: 9500 Gilman Drive, La Jolla, CA

92093, USA. Tel: 858-822-5771. Fax: 858-534-2005. Email: [email protected]

Keywords: zebrafish, apolipoprotein C-II, lipoprotein lipase, hyperlipidemia

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

http://dmm.biologists.org/lookup/doi/10.1242/dmm.019836Access the most recent version at DMM Advance Online Articles. Posted 4 June 2015 as doi: 10.1242/dmm.019836http://dmm.biologists.org/lookup/doi/10.1242/dmm.019836Access the most recent version at

First posted online on 4 June 2015 as 10.1242/dmm.019836

Page 2: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Summary Statement

Apoc2 loss-of-function zebrafish display severe hypertriglyceridemia, characteristic

of human patients with defective lipoprotein lipase activity. Use of apoc2 mutant

zebrafish will facilitate mechanistic studies and drug screening to treat

hypertriglyceridemia.

Abstract

Apolipoprotein CII 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 apoC-II mimetic peptide. Consistent with

the 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

pathogenesis of associated human diseases.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 3: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

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), while severe hypertriglyceridemia (TG > 885 mg/dL) is usually caused by

single gene mutations encoding lipoprotein lipase (LPL), or less commonly, 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 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; O'Hare 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 (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 (Gene ID: 568972, molecular

mass 11.2 kDa) is only 27.8% identical to human APOC2, with 49.5% conserved

consensus amino acids, but its C-terminal region (residues 67-92, zebrafish

numbering), which mediates Apoc2 binding to Lpl, shows 46.1% identity to the LPL-

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 4: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

binding region of human APOC2. Thus, it is likely that the APOC2/LPL complex

required for TG hydrolysis is also conserved in zebrafish. To support this hypothesis,

we have developed an Apoc2 loss-of-function zebrafish model. Our results

demonstrate that the apoc2 mutant zebrafish develop severe hypertriglyceridemia,

characteristic for human patients deficient in APOC2, and that the apoc2 mutant is a

suitable animal model to study hyperlipidemia and the mechanisms involved in the

pathogenesis of associated diseases.

Results

Mutation of zebrafish apoc2 gene with TALENs

To create a zebrafish model of hypertriglyceridemia, we mutated the apoc2

gene in zebrafish using a transcription activator-like effector nuclease (TALEN)

technique. We chose a target site located at exon 3 of the apoc2 gene, with TALEN

bindings sequences of 16 bp and 17 bp nucleotides on the left and right side of the

target site, respectively. The spacer DNA was 21 bp long and contained the BtsI

restriction enzyme site (Fig. 1A). The corresponding protein coding region of the

TALEN target site is located in front of the Lpl binding domain (Fig. 1B). The

mRNAs encoding TALENs were injected into one-cell stage zebrafish embryos. To

test whether the apoc2 TALEN pair disrupted the apoc2 gene at its specific target site,

we extracted genomic DNA from F0 generation zebrafish, amplified the regions

containing the target site with PCR and conducted BtsI enzyme digestion. Compared

to wild type (WT), a part of the PCR band amplified from TALEN-injected F0 was

resistant to enzyme digestion (Fig. 1C), consistent with modification of the BtsI

recognition site in the apoc2 genomic sequence. Then, F0 zebrafish were out-crossed

with wild type zebrafish and the F1 progeny were genotyped and outcrossed again for

two generations, followed by an incross to obtain homozygous mutants (Fig. 1D).

Sequencing of the apoc2 mutant revealed a one-nucleotide replacement and 10-

nucleotide deletion, resulting in a frame 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 qPCR with the primers specific to both WT

and mutated apoc2 mRNA. The apoc2 mRNA was undetectable in adult liver of the

mutant (Fig. 1F) and dramatically decreased in 5.5 dpf larvae (Fig. 1G). These data

suggest elimination of apoc2 mRNA transcripts that contain frame shift, as it has been

demonstrated for many other premature stop codon and frame shift transcripts (Baker

and Parker, 2004), and loss of Apoc2 function.

Hyperlipidemia in adult apoc2 mutants

Adult apoc2 mutants were viable and fertile, and had normal physical

appearance (Fig. 2A and supplementary material Fig. S1). To test whether apoc2

deficiency resulted in hypertriglyceridemia, we drew blood from adult male zebrafish

into micro-capillary tubes 8 hours after last feeding and let the capillaries stand

overnight at 4°C to sediment erythrocytes. Plasma from an apoc2 mutant showed a

milky phenotype, with a creamy layer at the top (Fig. 2B), typical of that found in

subjects with APOC2 or LPL deficiency (Baggio et al., 1986; Fellin et al., 1983).

These plasma samples were also subjected to native agarose gel electrophoresis.

Compared to WT, mutant plasma showed a decrease in HDL and a dramatic increase

in the VLDL/LDL fraction. Chylomicrons, which were never observed in fasted WT

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 5: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

zebrafish, were present in the mutant (Fig. 2C, gel origin). Size exclusion

chromatography of pooled zebrafish plasma showed a dramatic increase of TG and

total cholesterol (TC) in chylomicron/VLDL and LDL fractions, but decreased TC in

the HDL fraction in the plasma of apoc2 mutants compared to WT zebrafish (Fig. 2D

and E). For comparison, plasma from an Ldlr-/- mouse fed a 60% high-fat diet showed

a similar to apoc2 mutant plasma lipoprotein profile, albeit with lower TG levels.

Pooled plasma from overnight fasted zebrafish was also subjected to

ultracentrifugation to float triglyceride rich lipoproteins (TRL). The uppermost layer

corresponding to TRL, collected from WT and apoc2 mutant zebrafish, was then

subjected to electron microscopy to visualize lipoproteins. While no TRL were

detected in the WT plasma, TRL of different sizes, including large chylomicrons,

were abundantly present in apoc2 mutant plasma (Fig. 2F). Accordingly, plasma TG

levels were dramatically increased (2,488 ± 160 mg/dL vs. 67 ± 8 mg/dL, mutant vs.

WT; p < 0.0001, Fig. 2G) and plasma lipase activity was significantly decreased in

apoc2 mutants compared to WT (Fig. 2H). Although it has been reported that human

patients with APOC2 deficiency have a mild elevation in plasma cholesterol (Baggio

et al., 1986; Fellin et al., 1983), unexpectedly, plasma TC levels were increased as

much as 4-fold in apoc2 mutants compared to WT zebrafish (1,202 ± 63 mg/dL vs.

296 ± 26 mg/dL, mutant vs. WT; p < 0.001) (Fig. 2I).

Taken together, these data suggest that the TALEN-induced mutation in the

apoc2 gene resulted in severe hypertriglyceridemia and chylomicronemia, confirming

that the function of APOC2 to activate LPL is conserved in zebrafish.

Hyperlipidemia in apoc2 mutant larvae

apoc2 mutant embryos and larvae did not show significant defects in

development, other than minor but statistically significant delays in yolk utilization

and growth. At 30 hours post fertilization (hpf) and 3.5 dpf, the mutant’s yolk was

larger but the body length was shorter than that of the WT (Fig. 3 A-G). However, the

developmental stages at both 30 hpf and 3.5 dpf, which were determined by the head

position and the pigment patterns, were similar (Fig. 3 A and D).

It has been reported that apoc2 transient knockdown inhibits angiogenesis

(Avraham-Davidi et al., 2012). To assess embryonic angiogenesis, we crossed the

apoc2 mutant with fli1-EGFP transgenic zebrafish, which express EGFP in

endothelial cells. At 26 hpf, there were no angiogenesis differences between apoc2

mutant and WT zebrafish (Fig.3 G, a-a’). However, at 52 hpf, growth of inter-

segmental blood vessels (ISV) was delayed in the apoc2 mutant (Fig.3 G, b-b’), as

was the outgrowth of subintestinal veins (SIV) at 80 hpf (Fig.3 G, c-c’), consistent

with the results reported by Avraham-Davidi et al. Interestingly, these ISV and SIV

defects in mutants were all corrected at later stages and the vasculature of 14 dpf

mutants was normal (supplementary material Fig. S2).

For up to 5 dpf, zebrafish embryos, unlike adults, receive nutrients through

yolk utilization. To test whether TG and cholesterol levels were affected at this early

stage, which is a suitable time window for drug screening and genetic manipulation

by antisense morpholino oligonucleotides injection, we stained 1-6 dpf embryos with

Oil Red O (ORO), which stains neutral lipids, including TG and cholesterol esters

(CE). Before the 3rd dpf, the strongest ORO staining was in the yolk and no

significant differences between WT and apoc2 mutants were detected. Starting from 3

dpf, we noticed consistently stronger vascular ORO staining in the mutants, with the

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 6: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

most significant differences between WT and apoc2 mutants observed in non-fed 6

dpf embryos. At this stage, ORO staining was weak or absent in the blood vessels of

WT zebrafish, but strong ORO staining was detected in the apoc2 mutants (Fig. 4A,

a-a’, b-b’). The dramatic differences in the levels of circulating neutral lipids were

also monitored with the fluorescent dye BODIPY (Fig. 4A, c-c’). The advantage of

using BODIPY is that live zebrafish can be immersed in the stain and after a wash,

changes in neutral lipid levels can be monitored and quantified (Fig. 4B). Furthermore,

the BODIPY staining in live zebrafish persisted over a prolonged period of time (up

to 48 hours in apoc2 mutants in our experiments). Consistent with the staining results,

TG and TC levels were significantly increased in the homogenates of 6 dpf apoc2

mutant larvae compared to WT (Fig. 4C-D).

In addition to the apoc2 mutant characterized in Figs. 1-4, we generated a

second zebrafish line with a different apoc2 mutation, which introduced a stop codon

in the target site, causing loss of Lpl binding domain of Apoc2. This apoc2 mutant

(line 2) also displayed 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 functional conservation of Apoc2 in zebrafish.

Injection of normal plasma rescues hyperlipidemia in apoc2 mutants

In human patients with APOC2 deficiency, transfusions of normal plasma

result in rapid and dramatic decreases in patients’ plasma TG levels (Breckenridge et

al., 1978; Nordestgaard et al., 1988). To test whether this effect can be reproduced in

our apoc2 mutant zebrafish, we isolated plasma from adult WT and mutant zebrafish

and transfused them into WT and apoc2 mutant larva recipients through cardinal vein

injection. The plasma was supplemented with red fluorescent dextran as a tracer to

confirm a successful injection. Twenty-four hours after injection, the larvae with

positive dextran fluorescence were selected and stained with BODIPY. The WT

larvae injected with WT or mutant plasma did not show changes of BODIPY staining.

Compared to apoc2 mutant plasma-injected mutant larvae, WT plasma-injected apoc2

mutant larvae showed a significant decrease in BODIPY fluorescence (Fig. 5A and B).

Following BODIPY imaging, the same larvae were euthanized, homogenized and

lipids were measured. Consistent with the BODIPY staining results, TG levels in the

apoc2 mutants were significantly reduced following the injection of WT plasma (Fig.

5C and D).

Injection of apoC-II mimetic peptide rescues hyperlipidemia in apoc2 mutants

We have recently developed a novel bihelical amphipathic peptide (C-II-a)

that contains an amphipathic helix (18A) for binding to lipoproteins and stimulating

cholesterol efflux as well as a motif based on the last helix of human apoC-II that

activates lipolysis by LPL (Amar et al., 2015). An inactive apoC-II peptide (C-II-i) in

which 4 amino acids were mutated, was used as a negative control. The C-II-a peptide

restored normal lipolysis in plasma from apoC-II deficient human patients and

dramatically decreased the TG levels in Apoe-/- mice (Amar et al., 2015). Protein

alignment indicates that 9 of 20 amino acids in C-II-a are conserved between

zebrafish and human, including the critical amino acids that were mutated in C-II-i

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 7: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

(Fig. 6A). To test whether the human apoC-II-based mimetic peptide can rescue

hypertriglyceridemia in apoc2 mutant zebrafish, we injected C-II-a or C-II-i, together

with a fluorescent dextran tracer, into zebrafish larvae and assessed vascular content

of neutral lipids (using BODIPY staining) 30 min and 6, 30 and 72 hours after

injection. While at 30 min there was no difference, at 6 hours post C-II-a injection,

there was a modest, but statistically significant, 20% decrease in BODIPY

fluorescence (supplementary material Fig. S4). Further, at 30 hours, the BODIPY

signal was dramatically decreased – by 60% – in C-II-a, but not C-II-i injected

zebrafish (Fig. 6B and C), and the effect of C-II-a peptide persisted for at least 72

hours (supplementary material Fig. S4). Biochemical measurements confirmed a

significant decrease in TG levels and a trend toward TC decrease (Fig. 6D and E).

Vascular lipid accumulation in apoc2 mutant larvae

In our previous experiments, we found that supplementing a high-cholesterol

diet (HCD) with red fluorescent CE enabled monitoring accumulation of vascular

lipid deposits in live, transparent zebrafish larvae (Fang et al., 2011; Stoletov et al.,

2009). As apoc2 mutant zebrafish have higher TG and TC levels even when fed a diet

with normal content of cholesterol (Fig. 2, Fig. 4 and Fig. 7A and B), we tested the

apoc2 mutants for the presence of vascular lipid deposits. WT and apoc2 mutants

were fed a diet with normal content of fat and cholesterol from 5 to 14 dpf. To trace

lipid accumulation, fluorescently labeled CE (576/589-CE) was added to the diet from

8 to 12 dpf. The number of fluorescent lipid deposits was significantly higher in the

vasculature of apoc2 mutants than in WT zebrafish (Fig. 7C and D). To test whether

these vascular lipid deposits were in fact intracellular lipid accumulated in

macrophage, as observed in human and mouse early atherosclerotic lesions, we

crossed apoc2 mutants to mpeg1-EGFP transgenic zebrafish, which express EGFP in

macrophages. As expected, the majority of the lipid deposits were localized within

macrophages (Fig. 7E and F).

Pancreatic ectopic growth in apoc2 mutants

Patients with extremely high TG levels are at greater risk for development of

acute pancreatitis (Ewald et al., 2009). However, the mechanisms by which this

occurs are not known with certainty, but may be related in part to excess TG

accumulation in the pancreatic vasculature, where they would be subjected to

hydrolysis by pancreatic lipases (Wang et al., 2009). To assess developmental

pancreatic defects, we crossed apoc2 mutants with ptf1a-EGFP transgenic zebrafish,

which express EFGP primarily in acinar cells (Dong et al., 2008; Godinho et al.,

2005). We observed ectopic outgrowth of the acinar pancreas in apoc2 mutants

starting at 4 dpf, which was persistent and was observed in 6.5 dpf embryos as well

(Fig. 8A and B). At 6.5 dpf, a WT pancreas usually has a head domain, which

contains the principal islet and a long tail domain that extend posteriorly. In contrast,

the apoc2 mutant pancreas had additional short ectopic protrusions along the head of

the pancreas (Fig. 8). These defects found in embryos from apoc2-/- incrosses were

confirmed in apoc2+/- incross experiments in which embryos were scored for ectopic

pancreatic outgrowth prior to genotyping (Fig. 8C).

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 8: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Discussion

Recent genome-wide association studies provide new strong evidence and

support earlier studies suggesting that abnormal metabolism of TRL contributes to

increased risk of CVD (Do et al., 2015; Do et al., 2013; Hokanson and Austin, 1996).

In addition, it has been shown that carriers of APOC3 loss-of-function mutations have

40% lower plasma triglyceride levels and a 40% lower risk of CVD than the general

population (Jorgensen et al., 2014; Tg et al., 2014). These two Mendelian

Randomization studies categorized participants based on their APOC3 genotype

rather than plasma TG levels, theoretically excluding confounding factors that affect

both plasma TG levels and CVD (Cohen et al., 2014). Although one still cannot

exclude that APOC3 plays its role in CVD through other pathways independent of its

effects on TG levels, those two studies clearly link the plasma TG levels with the

genetic causality of CVD risk.

A major mechanism by which APOC3 increases plasma TG is by its ability to

inhibit LPL activity (Larsson et al., 2013), although it also is known to inhibit the

clearance of VLDL and chylomicron remnants. In contrast, APOC2 is a cofactor of

LPL and its loss of function results in deficient LPL activity and consequent

hypertriglyceridemia 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 in the apoc2 gene results in chylomicronemia and profound

hypertriglyceridemia. The first apoc2 mutant we selected had a frame shift mutation,

which led to the amino acid sequence change in its Lpl binding domain (Fig. 1B and

E). In a different apoc2 mutant line, we found a stop codon mutation, which caused

deletion of the Lpl binding domain from the Apoc2 protein sequence (supplementary

material Fig. S3). Importantly, both apoc2 mutant zebrafish lines showed dramatically

decreased apoc2 mRNA expression and developed hypertriglyceridemia, validating

the Apoc2 loss-of-function model.

Plasma of adult apoc2 mutant zebrafish have many characteristics common

with the plasma of APOC2 or LPL deficient human patients (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). The advantages of using

zebrafish as an animal model include the optical transparency of embryos and larvae,

which makes them suitable for live, high-resolution imaging, and easy genetic

manipulation and drug screening. Our studies demonstrate that as early as at 3 dpf

apoc2 mutant larvae already display remarkable hyperlipidemia, and in 6 dpf embryos,

dramatic differences in circulating neutral lipids were detected with ORO staining and

confirmed with biochemical measurements of TG and TC levels in whole body

homogenates (Fig. 4).

In human APOC2 patients, chylomicron and VLDL size and abundance

fluctuate widely and depend on the type of food consumed on the preceding day

(Breckenridge et al., 1978). Future work will determine if variation of a diet fed to

apoc2 mutant zebrafish will help maximize chylomicron or VLDL content for

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 9: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

specific experimental goals. An important difference between mammalian and fish

lipoprotein metabolism is the absence of APOBEC1 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 of human chylomicrons. This may explain

that the majority of TRL we observed in apoc2 mutant zebrafish have the size similar

to human and mouse VLDL. The lipoprotein profiles shown in Fig. 2D-F suggest that

the apoc2 mutant zebrafish are suitable for modeling human dyslipidemia, and the

zebrafish model favorably compares to a large number of other models, including the

Ldlr-/- mouse, profiled in (Yin et al., 2012).

In this work, we developed a simple and convenient protocol for staining

circulating lipoproteins with BODIPY. BODIPY is a lipophilic fluorescent dye, which

preferentially localizes to the neutral lipid core of lipoproteins and intracellular lipid

droplets, and is widely used in cell biology (Bozaquel-Morais et al., 2010; Gocze and

Freeman, 1994; Grandl and Schmitz, 2010). However, to the best of our knowledge,

BODIPY was not used to stain and monitor circulating lipid levels in live zebrafish

larvae. In our experiments, live zebrafish were immersed in a BODIPY solution, with

a subsequent wash in water. This simple procedure resulted in bright green

fluorescent staining of neutral lipids in the circulation of apoc2 mutant animals. This

assay provides a robust readout and may help conduct genetic and drug screening

studies.

We used the BODIPY assay to demonstrate that injections of normal zebrafish

plasma significantly diminished hypertriglyceridemia in apoc2 mutant zebrafish (Fig.

5). This result is similar to the treatment of APOC2 deficient patients with severe

hypertriglyceridemia in which transfusions of normal, APOC2-containing plasma led

to rapid, within one day, reductions in plasma triglycerides (Breckenridge et al., 1978).

Remarkably, the amino acids critical for LPL activation in the last helix of human

APOC2 are conserved in zebrafish, and the human apoC-II mimetic peptide C-II-a

(Amar et al., 2015), rescued severe hypertriglyceridemia in apoc2 mutant zebrafish

(Fig. 6). These results suggest that our zebrafish model is suitable for testing potential

therapeutic agents for treatment of human APOC2 deficiency.

Two previous studies in which apoc2 was transiently knocked down in

zebrafish embryos with antisense morpholino oligonucleotides, suggested that Apoc2

is involved in yolk absorption and angiogenesis (Avraham-Davidi et al., 2012; Pickart

et al., 2006). Our results with the apoc2 mutant zebrafish support these conclusions.

Consistent with the results reported by Pickart et al., apoc2 mutant larvae show a

slightly delayed yolk consumption (Fig.3 A-G) and the mutant larvae are of smaller

size when compared to WT larvae. This phenotype in apoc2 mutant can be explained,

in part, by the disruption of the Lpl function, resulting in a defect in TG hydrolysis

and supply of FFA to non-hepatic tissues, which, in turn, may cause nutrient

deprivation and delayed growth. On the other hand, by monitoring the head angle at

28 hpf and the pattern of pigmentation at 3 dpf, we did not notice any differences in

the stage development between WT and apoc2 mutants. Because adult apoc2 mutant

zebrafish are of the same size as WT and have no apparent phenotype (Fig.2A and

supplementary material Fig. S1), other than hyperlipidemia, it is possible that feeding

and the activity of other lipases help restore the energy balance in unchallenged

animals.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 10: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

A previous study reported that a transient apoc2 knockdown by antisense

morpholino oligonucleotides increased levels of apoB containing lipoproteins, which,

in turn, inhibited embryonic angiogenesis (Avraham-Davidi et al., 2012). In our

experiments, the Apoc2 loss of function resulted in dramatically increased levels of

VLDL, an apoB containing lipoprotein, and indeed delayed ISV and SIV growth.

Nevertheless, the apoc2 mutants are viable and their vascular structure is normal at 14

dpf despite profound hyperlipidemia (supplementary material Fig. S2). Thus, the

excess of apoB containing lipoproteins is likely to delay but not to completely inhibit

angiogenesis in vivo.

In agreement with epidemiologic studies suggesting that hypertriglyceridemia

is a risk factor for CVD (Do et al., 2015; Jorgensen et al., 2014; Tg et al., 2014), our

results demonstrate that the hypertriglyceridemia induced by the apoc2 mutation in

zebrafish results in vascular lipid accumulation and macrophage lipid uptake (Fig. 7

C-E), which are important initial events in development of human atherosclerosis.

The vascular lipid accumulation in zebrafish induced by hypertriglyceridemia was

similar to that induced by hypercholesterolemia (O'Hare et al., 2014; Stoletov et al.,

2009). Our study is also consistent with mouse studies in which hypertriglyceridemia

induced by Lpl or Gpihbp knockout resulted in spontaneous development of

atherosclerosis (Weinstein et al., 2010; Zhang et al., 2008). In contrast to

hypertriglyceridemic mice in which atherosclerotic lesions develop late, by the age of

1 year, vascular lipid deposits in apoc2 mutant zebrafish fed normal diet develop as

early as at 14 dpf. One possible explanation is the absence of CETP in mice, whereas

zebrafish express functional Cetp (Jin et al., 2011; Kim et al., 2012). Transfer of

cholesterol from HDL to LDL and VLDL (and TG in the opposite direction) may play

an atherogenic role in apoc2 mutant zebrafish, as it does in humans.

One important clinical complication of hypertriglyceridemia in patients with

LPL or APOC2 deficiency is recurrent pancreatitis, but the underlying mechanisms

are poorly defined (Baggio et al., 1986; Fellin et al., 1983). In apoc2 mutant zebrafish,

we found ectopic acinar outgrowth in the head of the pancreas (Fig. 8). Acinar cells

express high levels of pancreatic lipase. Chylomicronemia results in increased FFA

uptake and injury in the acinar cells, which may lead to inflammation and pancreatitis

(Scherer et al., 2014; Wang et al., 2009). Our findings suggest that the Apoc2

deficiency may affect early pancreas development in zebrafish. A recent report shows

that modeling chylomicron retention disease in zebrafish, which manifests in the

reduction of circulating neutral lipids, was associated with inhibited growth of

exocrine pancreas (Levic et al., 2015). We yet do not know whether the hyper- or

hypo-triglyceridemia effects on exocrine pancreas growth in apoc2 mutant zebrafish

are related to the pathogenesis 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 animal model to study mechanisms

related to human diseases induced by hypertriglyceridemia. Small molecule and

genetic screens using the apoc2 mutant zebrafish may suggest new approaches to

treatment of hyperlipidemia and related disorders.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 11: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Materials and Methods

Zebrafish maintenance and feeding. Adult zebrafish of the AB strain were

maintained at 28°C on a 14-hour-light/10-hour-dark cycle and fed brine shrimp twice

a day. Zebrafish larvae were fed Golden Pearls (100-200 m size from Brine Shrimp

Direct, Ogden, UT) twice a day, starting from 4.5 dpf. For lipid deposition

experiments, Golden Pearls, supplemented with 1 g/g of a fluorescent cholesterol

ester analog (cholesteryl BODIPY 576/589-C11 from Invitrogen, Carlsbad, CA;

C12681), were fed to zebrafish from 8 to 12 dpf as described (Stoletov et al., 2009).

fli1:EGFP and mpeg1-EGFP transgenic zebrafish were from the Weinstein (Lawson

and Weinstein, 2002) and Lieschke (Ellett et al., 2011) laboratories, respectively.

ptf1a-EGFP transgenic zebrafish were developed in the Dong lab (Dong et al., 2008;

Godinho et al., 2005). All animal studies were approved by the UCSD Institutional

Animal Care and Use Committee.

TALEN construction, mRNA synthesis, injection and confirmation of apoc2

mutant. TALEN plasmids targeting apoc2 were constructed from the stock cassette

according to the published protocols (Dong et al., 2011; Huang et al., 2014). TALEN

mRNAs were synthesized with mMESSAGE mMACHINE Sp6 Transcription kit

(Ambion, Austin, TX; AM1340). mRNAs encoding the pair of TALEN proteins were

mixed at a 1:1 ratio to a final concentration of 200 ng/l. One-to-two nl of TALEN

mRNAs was injected into the one-cell stage embryos. Genomic DNA (gDNA) were

extracted from whole embryos or from the tissue clipped off tail fin of adult zebrafish

and were genotyped by PCR (with the following primers: forward,

GTGGTCGCATAGTTTAAGCT; reverse, GCAACCATGTAAGAGTTGCA) and

BtsI enzyme digestion, and confirmed by sequencing, according to published

protocols (Dong et al., 2011; Huang et al., 2014). To assess expression of apoc2 in

larvae homogenates and in adult liver, total RNA was isolated, reverse transcribed and

subjected to qPCR with the following primers: forward,

ATGAACAAGATACTGGCTAT; reverse, TTGATGGTCTCTACATATCC, using a

KAPA SYBR FAST Universal qPCR kit (KAPA Biosystems, Wilmington, MA;

KK4602) and a Rotor Gene Q qPCR machine (Qiagen, Valencia, CA). The position

of these qPCR primers relative to the TALEN target site is shown in Fig. S3. Because

the mutation does not change nucleotide sequence beyond the target site, these

primers detect expression of both WT and mutant mRNA, if any is present.

Oil Red O staining and BODIPY staining. Oil red O (ORO) staining was conducted

according to published protocols (Schlegel and Stainier, 2006). Briefly, embryos were

fixed in 4% paraformaldehyde (PFA) for 2 hours, washed 3 times in PBS, incubated

in 0.3% ORO solution for 2 hours and then washed with PBS before imaging. For

BODIPY 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 g/ml BODIPY® 505/515

(4,4-Difluoro-1,3,5,7-Tetramethyl-4-Bora-3a,4a-Diaza-s-Indacene, Invitrogen, D-

3921) for 1 hour in dark and then rinsed with E3 water before imaging.

Triglyceride and cholesterol measurements and lipoprotein analysis. Blood was

collected from adult male zebrafish, after overnight fasting, through tail amputation

and diluted 1:50 (WT) or 1:200 (apoc2 mutant) in PBS. The supernatants were

collected as a plasma fraction after centrifugation at 2,350 g for 10 minutes. Embryos

or larvae were gently homogenized in PBS with pestle. After centrifugation at 16,000

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 12: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

g for 10 minutes, supernatants were collected and referred to as “homogenates.” TG

and TC levels were measured in 25 l of diluted plasma or embryo/larva homogenates

using kits from BioVision (Milpitas, CA. Triglyceride Quantification Kit: K622-100;

Cholesterol Quantification Kit: K623-100) and according to manufacturer’s protocol.

To assess undiluted plasma, 5 l blood was collected in a tube containing 0.5 l

heparin (5 mg/ml) and then loaded into a heparin rinsed capillary, which was kept

overnight at 4°C in a vertical position. Following erythrocyte sedimentation, the

capillaries were photographed. Lipoprotein fractions were assessed using native

agarose gel electrophoresis (Helena Laboratories, Beaumont, TX; 3045) as we

previously described (Stoletov et al., 2009).

FPLC lipoprotein profile. One hundred l of pooled plasma from 45 adult WT

zebrafish or 40 l of pooled plasma from 14 adult apoc2 mutant zebrafish were

loaded onto a Superose 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 microscopy. Pooled plasma from fasted overnight

WT and apoc2 mutant zebrafish were diluted in PBS, without density adjustment, and

the samples were centrifuged at 200,000 g for 4 hours at 4 °C. The top layer

containing TRL was collected, supplemented with 0.1% sucrose, added to a FCF100-

Cu 100 mesh (Electron Microscopy Sciences, Hatfield, PA) and negatively stained

with 1% uranyl acetate. Stained samples were imaged with a Tecnai G2 Spirit

BioTWIN transmission electron microscope equipped with a 4K Eagle digital camera

(FEI Company, Hillsboro, OR).

Plasma lipase activity assay. Adult zebrafish blood was diluted 1:25 in PBS and

after centrifugation, 5 l of the diluted plasma was used to measure lipase assay with

a Lipase Activity Assay Kit (Cayman Chemical, Ann Arbor, MI; 700640), following

the manufacturer’s manual. The assay buffer was used as a negative control. The

reaction was conducted at 30°C.

Intravenous injections. Blood was collected by tail amputation from adult male

zebrafish 4 hours after last feeding, and plasma was separated by centrifugation at

2,350 g for 10 minutes. 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 of successful

injection, zebrafish plasma or apoC-II mimetic peptides were supplemented with 2%

(v/v) tetramethylrhodamine-conjugated 10 kDa dextran (0.5 mg/ml miniRuby,

Invitrogen, D-3312). For injections, 5 dpf larvae were aligned in 0.5% low melting

point agarose. Five nl of plasma or apoC-II mimetic peptides were injected through

the cardinal vein above the heart chamber using a FemtoJet microinjector (Eppendorf,

Hamburg, Germany). The red signal from fluorescent dextran confirmed successful

injection.

Imaging of live embryos or larvae. For in vivo live microscopy, anesthetized

embryos or larvae were mounted in low melting 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 were captured

with Leica CTR5000 (Wetzlar, Germany), Olympus FV1000 spectral confocal

(Tokyo, Japan) or BZ9000 Keyence (Osaka, Japan) fluorescent microscopes.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 13: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Competing Interests

J.L.W. is a consultant for ISIS Pharmaceuticals. Other authors declare no competing

of interests.

Author contributions

C.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. and

P.D.S.D. provided critical materials, ideas and discussion for this work. C.L. and

Y.I.M. wrote the paper.

Funding

The project was supported by the NIH grants HL093767 (Y.I.M.), HL055798 (Y.I.M.)

and HL088093 (J.L.W.), DK098092 (P.D.S.D.), as well as the UCSD Neuroscience

Microscopy Facility Grant P30NS047101.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 14: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

References

Amar, M. J., Sakurai, T., Sakurai-Ikuta, A., Sviridov, D., Freeman, L., Ahsan, L. and Remaley, A. T. (2015). A Novel Apolipoprotein C-II Mimetic Peptide That Activates Lipoprotein Lipase and Decreases Serum Triglycerides in Apolipoprotein E-Knockout Mice. J Pharmacol Exp Therapeut 352, 227-35. 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, 111-41. 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 modulating expression of VEGF receptor 1. Nat Med 18, 967-73. 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, lipase activity, and correction of hypertriglyceridemia after apolipoprotein C-II administration in two affected patients. J Clin Invest 77, 520-7. Baker, K. E. and Parker, R. (2004). Nonsense-mediated mRNA decay: terminating erroneous gene expression. Curr Opin Cell Biol 16, 293-9. Bozaquel-Morais, B. L., Madeira, J. B., Maya-Monteiro, C. M., Masuda, C. A. and Montero-Lomeli, M. (2010). A new fluorescence-based method identifies protein phosphatases 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, 1265-73. Cohen, J. C., Stender, S. and Hobbs, H. H. (2014). APOC3, Coronary Disease, and Complexities of Mendelian Randomization. Cell Metab 20, 387-9. Conticello, S. G., Thomas, C. J., Petersen-Mahrt, S. K. and Neuberger, M. S. (2005). Evolution of the AID/APOBEC family of polynucleotide (deoxy)cytidine deaminases. Mol Biol Evol 22, 367-77. Cox, D. W., Breckenridge, W. C. and Little, J. A. (1978). Inheritance of apolipoprotein C-II deficiency with hypertriglyceridemia and pancreatitis. N Engl J Med 299, 1421-4. Do, R., Stitziel, N. O., Won, H. H., Jorgensen, A. B., Duga, S., Angelica Merlini, P., Kiezun, A., Farrall, M., Goel, A., Zuk, O. et al. (2015). Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature 518, 102-6. 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 variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet 45, 1345-52. Dong, P. D., Provost, E., Leach, S. D. and Stainier, D. Y. (2008). Graded levels of Ptf1a differentially regulate endocrine and exocrine fates in the developing pancreas. Genes Dev 22, 1445-50. 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 human disease. Curr Opin Genet Dev 10, 252-6. Ebara, T., Ramakrishnan, R., Steiner, G. and Shachter, N. S. (1997). Chylomicronemia due to apolipoprotein CIII overexpression in apolipoprotein E-null mice.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 15: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Apolipoprotein CIII-induced hypertriglyceridemia is not mediated by effects on apolipoprotein E. J Clin Invest 99, 2672-81. Ellett, F., Pase, L., Hayman, J. W., Andrianopoulos, A. and Lieschke, G. J. (2011). mpeg1 promoter transgenes direct macrophage-lineage expression in zebrafish. Blood 117, e49-56. Ewald, N., Hardt, P. D. and Kloer, H. U. (2009). Severe hypertriglyceridemia and pancreatitis: 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 and attenuation of oxidized lipid accumulation in hypercholesterolemic zebrafish. J Clin Invest 121, 4861-9. 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 lipase and apolipoprotein C-II deficiency. Lipoprotein and apoprotein analysis, adipose tissue and hepatic lipoprotein lipase levels in seven patients and their first degree relatives. Atherosclerosis 49, 55-68. Fukushima, N. and Yamamoto, K. (2004). [Apolipoprotein C-I, C-II, C-III]. Nihon Rinsho 62 Suppl 12, 110-3. 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 than white fat biology. J Biol Chem 288, 14046-58. Gocze, P. M. and Freeman, D. A. (1994). Factors underlying the variability of lipid droplet fluorescence in MA-10 Leydig tumor cells. Cytometry 17, 151-8. Godinho, L., Mumm, J. S., Williams, P. R., Schroeter, E. H., Koerber, A., Park, S. W., Leach, S. D. and Wong, R. O. (2005). Targeting of amacrine cell neurites to appropriate synaptic laminae in the developing zebrafish retina. Development 132, 5069-79. Goldberg, I. J., Eckel, R. H. and McPherson, R. (2011). Triglycerides and heart disease: still a hypothesis? Arterioscler Thromb Vasc Biol 31, 1716-25. Goldberg, I. J. and Merkel, M. (2001). Lipoprotein lipase: physiology, biochemistry, and molecular biology. Front Biosci 6, D388-405. Goulbourne, C. N., Gin, P., Tatar, A., Nobumori, C., Hoenger, A., Jiang, H., Grovenor, C. R., Adeyo, O., Esko, J. D., Goldberg, I. J. et al. (2014). The GPIHBP1-LPL complex is responsible for the margination of triglyceride-rich lipoproteins in capillaries. Cell Metab 19, 849-60. Grandl, M. and Schmitz, G. (2010). Fluorescent high-content imaging allows the discrimination and quantitation of E-LDL-induced lipid droplets and Ox-LDL-generated phospholipidosis in human macrophages. Cytometry A 77, 231-42. Hokanson, J. E. and Austin, M. A. (1996). 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. J Cardiovasc Risk 3, 213-9. Holtta-Vuori, M., Salo, V. T., Nyberg, L., Brackmann, C., Enejder, A., Panula, P. and Ikonen, E. (2010). Zebrafish: gaining popularity in lipid research. Biochem J 429, 235-42. Hooper, A. J., Kurtkoti, J., Hamilton-Craig, I. and Burnett, J. R. (2014). Clinical features 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). TALEN construction via "Unit Assembly" method and targeted genome modifications in zebrafish. Methods 69, 67-75.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 16: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Jin, S., Hong, J. H., Jung, S. H. and Cho, K. H. (2011). Turmeric and laurel aqueous extracts exhibit in vitro anti-atherosclerotic activity and in vivo hypolipidemic effects in a zebrafish model. J Medic Food 14, 247-256. Jorgensen, A. B., Frikke-Schmidt, R., Nordestgaard, B. G. and Tybjaerg-Hansen, A. (2014). Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 371, 32-41. Kei, A. A., Filippatos, T. D., Tsimihodimos, V. and Elisaf, M. S. (2012). A review of the role of apolipoprotein C-II in lipoprotein metabolism and cardiovascular disease. Metabolism 61, 906-21. Kim, J. Y., Hong, J. H., Jung, H. K., Jeong, Y. S. and Cho, K. H. (2012). Grape skin and 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, 606-14. Larsson, M., Vorrsjo, E., Talmud, P., Lookene, A. and Olivecrona, G. (2013). Apolipoproteins C-I and C-III inhibit lipoprotein lipase activity by displacement of the enzyme from lipid droplets. J Biol Chem 288, 33997-4008. Lawson, N. D. and Weinstein, B. M. (2002). In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev Biol 248, 307-18. Levak-Frank, S., Hofmann, W., Weinstock, P. H., Radner, H., Sattler, W., Breslow, J. L. and Zechner, R. (1999). Induced mutant mouse lines that express lipoprotein 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 U S A 96, 3165-70. Levic, D. S., Minkel, J. R., Wang, W. D., Rybski, W. M., Melville, D. B. and Knapik, E. W. (2015). Animal model of Sar1b deficiency presents lipid absorption deficits similar to Anderson disease. J Mol Med (Berlin) 93, 165-76. 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. A mouse 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 the American Heart Association. Circulation 123, 2292-333. 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 Collaborative Lipid Research Clinics Family Study: biological and cultural determinants of familial resemblance for plasma lipids and lipoproteins. Genet Epidemiol 2, 227-54. Nordestgaard, B. G., Stender, S. and Kjeldsen, K. (1988). Reduced atherogenesis in cholesterol-fed diabetic rabbits. Giant lipoproteins do not enter the arterial wall. Arteriosclerosis 8, 421-8. O'Hare, E. A., Wang, X., Montasser, M. E., Chang, Y. P., Mitchell, B. D. and Zaghloul, N. A. (2014). Disruption of ldlr causes increased LDL-cholesterol and vascular lipid accumulation in a zebrafish model of hypercholesterolemia. J Lipid Res 55, 2242-2253. Okubo, M., Toromanovic, A., Ebara, T. and Murase, T. (2014). Apolipoprotein C-II : A novel large deletion in APOC2 caused by Alu-Alu homologous recombination in an 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: comprehensive expression 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

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 17: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

genetics framework to identify novel functions of the vertebrate secretome. PLoS One 1, e104. Scherer, J., Singh, V. P., Pitchumoni, C. S. and Yadav, D. (2014). Issues in hypertriglyceridemic pancreatitis: an update. J Clin Gastroenterol 48, 195-203. Schlegel, A. and Stainier, D. Y. R. (2006). Microsomal Triglyceride Transfer Protein Is Required for Yolk Lipid Utilization and Absorption of Dietary Lipids in Zebrafish Larvae. 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 transgenic mice. J Clin Invest 93, 1683-90. 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 lipid accumulation, lipoprotein oxidation, and macrophage lipid uptake in hypercholesterolemic zebrafish. Circ Res 104, 952-60. 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. et al. (2012). Mutations in LPL, APOC2, APOA5, GPIHBP1 and LMF1 in patients with severe hypertriglyceridaemia. J Intern Med 272, 185-96. Teng, B. and Davidson, N. O. (1992). Evolution of intestinal apolipoprotein B mRNA editing. Chicken apolipoprotein B mRNA is not edited, but chicken enterocytes contain in vitro editing enhancement factor(s). J Biol Chem 267, 21265-72. Tg, Hdl Working Group of the Exome Sequencing Project, N. H. L., Blood, I., Crosby, J., Peloso, G. M., Auer, P. L., Crosslin, D. R., Stitziel, N. O., Lange, L. A., Lu, Y. et al. (2014). Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 371, 22-31. Trent, C. M., Yu, S., Hu, Y., Skoller, N., Huggins, L. A., Homma, S. and Goldberg, I. J. (2014). Lipoprotein lipase activity is required for cardiac lipid droplet production. J Lipid Res 55, 645-58. 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 to pancreatitis in severe hypertriglyceridaemic lipoprotein lipase-deficient mice and agonist-like function of pancreatic lipase in pancreatic cells. Gut 58, 422-30. Watts, G. F., Ooi, E. M. and Chan, D. C. (2013). Demystifying the management of hypertriglyceridaemia. Nat Rev Cardiol 10, 648-61. 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). Chylomicronemia elicits atherosclerosis in mice--brief report. Arterioscler Thromb Vasc Biol 30, 20-3. 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 death in lipoprotein lipase knockout mice. Mild hypertriglyceridemia with impaired very low density lipoprotein clearance in heterozygotes. J Clin Invest 96, 2555-68. 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 optimal animal 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 in aged lipoprotein lipase-deficient mice with severe hypertriglyceridemia on a normal chow diet. Circ Res 102, 250-6.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 18: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figures

Figure 1: Generation of apoc2 mutant zebrafish

(A) The diagram of partial apoc2 genomic DNA sequence, showing the TALEN

binding sites (underlined) and the spacer DNA (lower case), which contains the BtsI

restriction site. (B) The diagram of the TALEN target site in the Apoc2 protein. (C)

Test for TALEN mutagenesis in F0 zebrafish with BtsI enzyme digestion. Arrow

indicates the modified genomic DNA, which was no longer recognized and cut by

BtsI. (D) Representative genotyping results of F2 generation. (E) Sequencing data

suggest a frame shift mutation at the site preceding the LPL binding domain in the

apoc2 mutant. (F and G) apoc2 mRNA expression (qPCR) in adult liver and 5.5 dpf

larvae. Mean±SEM; n=3 in each group. **, p<0.01.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 19: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figure 2: Adult apoc2 mutant zebrafish develop hyperlipidemia when fed normal

diet

(A) No apparent differences between 4 months old male WT and apoc2 mutant

zebrafish. Scale bar, 0.5 cm. (B) Clear and milky appearance of plasma from WT and

apoc2 mutant zebrafish, respectively. (C) Native gel electrophoresis and neutral lipid

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 20: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

staining of plasma from WT and apoc2 mutant zebrafish; CM, chylomicron. (D and E)

FPLC lipoprotein profile of pooled WT (45 animals) and apoc2 mutant (14 animals)

zebrafish plasma. Plasma from an Ldlr-/- mouse fed a 60% HFD was used for

comparison. (F) Electron microscopy images of TRL isolated from pooled WT and

apoc2 mutant zebrafish plasma by ultracentrifugation as described in Methods. (G)

Plasma TG levels. Mean±SEM; n=6 in each group; ***, p<0.001. (H) Plasma lipase

activity assayed as described in Methods. Assay buffer was used as negative control.

Mean±SEM; n=3 in each group; ***, p<0.001. (I) Plasma TC levels. Mean±SEM;

n=6 in each group; ***, p<0.001.

Figure 3: Development and angiogenesis in apoc2 mutant larvae

(A-C) Morphology of WT (n=11) and apoc2 mutant (n=11) embryos at 30 hpf.

Mean±SD; ***, p<0.001. (D-G) Morphology of WT (n=11) and apoc2 mutant (n=10)

embryos at 3.5 dpf. Mean±SEM; *, p<0.05; **, p<0.01; ***, p<0.001. Blue, red and

yellow lines delineate the size of yolk, yolk extension and the body length,

respectively. (H) Angiogenesis in fli1:EGFP WT and apoc2 mutant embryos at 28

hpf (a-a’), 52 hpf (b-b’) and 80 hpf (c-c’). The numbers in the figure indicate the

number of animals exhibiting the illustrated phenotype versus the total number of

animals. Scale bars in (A) and (D), 200 m; in (H), 100 m.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 21: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figure 4: apoc2 mutant larvae develop hyperlipidemia

(A) Non-fed 6 dpf WT and apoc2 mutant larvae were stained with Oil Red O (a-a’, b-

b’) or BODIPY (c-c’). Scale bar, 200 m. (B) Quantification of BODIPY staining in

panel A. Mean±SEM; n=3 in each group; ***, p<0.001. (C and D) TG and TC levels

in homogenates of 6 dpf WT and apoc2 mutant larvae, normalized to total protein

content. Mean±SEM; n=3 in each group; ***, p<0.001; **, p<0.01.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 22: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figure 5: Injections of WT zebrafish plasma rescue hyperlipidemia phenotype in

apoc2 mutants

(A) apoc2 WT or mutant larvae at 5 dpf were injected with 5 nl of undiluted plasma

from either WT or mutant adult male zebrafish, both supplemented with red

fluorescent dextran. One day after injection, at 6 dpf, larvae were stained with

BODIPY (green fluorescence) to assess neutral lipid content of plasma. Red dextran

fluorescence indicates successful injection. Scale bar, 100 m. (B) Quantification of

results shown in panel A. Mean±SEM; n=3 in each group; ***, p<0.001. (C and D)

TG and TC levels in homogenates of 6 dpf WT and apoc2 mutant larvae, normalized

to total protein content. Mean±SEM; five embryos pooled per sample, n=3 in each

group; **, p<0.01; *, p<0.05.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 23: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figure 6: Injections of human apoC-II mimetic peptide rescue hyperlipidemia

phenotype in apoc2 mutant

(A) Protein alignment of apoC-II mimetic peptides with the corresponding zebrafish

Apoc2 amino acids sequence. Blue and red asterisks indicate the conserved amino

acids in C-II-a (active) peptide; red asterisks indicate amino acids that were mutated

in C-II-i (inactive) peptide. (B) Thirty hours after injection of 5 nl of C-II-a or C-II-i,

at 6.5 dpf, larvae were stained with BODIPY. Red dextran fluorescence indicates

successful injection. Scale bar, 50 m. (C) Quantification of BODIPY staining results.

Mean±SEM; n=3 in each group; ***, p<0.001; ns, not significant. (D and E). TG and

TC levels in homogenates of 6.5 dpf WT and apoc2 mutant larvae, normalized to total

protein content. Mean±SEM; five embryos pooled per sample, n=3 in each group; **,

p<0.01; *, p<0.05.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 24: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figure 7: Enhanced vascular lipid accumulation in apoc2 mutants

(A and B) TG and TC levels in homogenates of 14 dpf WT and apoc2 mutant larvae,

normalized to total protein content. Larvae were fed normal diet. Mean±SEM; n=3 in

each group; ***, p<0.001; *, p<0.05. (C and D) Vascular lipid deposits in 14 dpf WT

(n=8) and apoc2 mutant (n=8) larvae fed normal diet; **, p<0.01. (E) Co-localization

of vascular lipid deposits with macrophages in 14 dpf apoc2 mutant mpeg1:EGFP

zebrafish. (F) Quantification of lipid deposits associated and non-associated with

macrophages in apoc2 mutant larvae. Mean±SEM; n=7. Scale bars in (C) and (E),

50m.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 25: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

Figure 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 apoc2 mutant larvae. Yellow arrow points to the normal pancreatic growth

and white arrows point to ectopic outgrowth. Scale bar, 100 m. (B and C) At 6.5dpf,

an independent observer scored a pancreatic outgrowth phenotype as 0=normal,

1=mild and 2=severe. (B) WT (n=13) and apoc2-/- mutant (n=21). Mean±SEM; ***,

p<0.001. (C) Heterozygous, apoc2+/- zebrafish carrying one copy of the ptf1a-EGFP

transgene were crossed with the heterozygous, apoc2+/- zebrafish carrying no ptf1a-

EGFP. Twenty-two embryos, which displayed EGFP fluorescence were selected,

scored for the pancreas 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). Mean±SEM; *, p<0.05; ***, p<0.001.

Translational Impact

Clinical issue: Blood transports dietary lipids and the lipids produced by the liver in

the form of large lipoprotein particles, such as chylomicrons and VLDL. These

lipoproteins also carry a protein, APOC2, which is necessary to activate lipoprotein

lipase (LPL) to hydrolyze triglycerides and deliver fatty acids to tissues.

Hypertriglyceridemia, arising from various factors, including LPL deficiency, is an

independent risk factor for cardiovascular disease and is positively associated with

metabolic disorders. Patients with a genetic defect in APOC2 or LPL have severe

hypertriglyceridemia, manifesting eruptive xanthomas, lipemia retinalis and acute and

recurrent pancreatitis, which can be lethal. However, mechanisms underlying

hypertriglyceridemia-associated diseases are not well understood, in part, due to

limited availability of animal models. Although substantial work has been conducted

with Lpl knockout mice, Apoc2 knockout mouse models have not been reported.

Results: Zebrafish is an emerging animal model to study lipid metabolism and

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 and cost-effective maintenance. In this work, Apoc2 loss-

of-function mutant zebrafish displayed chylomicronemia and severe

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt

Page 26: Apoc2 loss-of-function zebrafish mutant as a genetic model ... · 6/3/2015  · human LPL in either skeletal or cardiac muscle (Levak-Frank et al., 1999; Weinstock et al., 1995)

hypertriglyceridemia, which closely resembled the phenotype observed in human

patients with APOC2 deficiency. The hypertriglyceridemia in apoc2 mutants was

rescued by injection of plasma from wild type zebrafish with functional Apoc2 or by

injection of a human apoC-II mimetic peptide. apoc2 mutants fed a normal diet

accumulated lipid and lipid-laden macrophages in the vasculature, which resembles

early events in the development of human atherosclerotic lesions. In addition, apoc2

mutant embryos showed ectopic overgrowth of pancreas.

Implications and future directions: The new apoc2 mutant zebrafish display a robust

hyperlipidemia phenotype and could be a useful and versatile animal model to study

mechanisms related to human diseases induced by hypertriglyceridemia. Small

molecule and genetic screens using the apoc2 mutant zebrafish may suggest new

approaches to treatment of hyperlipidemia and related diseases.

Dise

ase

Mod

els &

Mec

hani

sms

D

MM

Acce

pted

man

uscr

ipt