Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Received: 9 November 2022 | Accepted: 30 December 2022

DOI: 10.1002/ame2.12309

ORIGINAL ARTICLE

Establishment of a non-­alcoholic fatty liver disease model by


high fat diet in adult zebrafish

Xiang Li1,2 | Lei Zhou2 | Yuying Zheng2 | Taiping He1 | Honghui Guo1 |
Jiangbin Li3 | Jingjing Zhang2

1
Department of Nutrition, School of Public
Health, Guangdong Medical University, Abstract
Zhanjiang, China
Background: Non-­alcoholic fatty liver disease (NAFLD) has become the most common
2
Zhanjiang Key Laboratory of Zebrafish
Model for Development and Disease,
chronic liver disease in recent years, but the pathogenesis is not fully understood.
Affiliated Hospital of Guangdong Medical Therefore, it is important to establish an effective animal model for studying NAFLD.
University, Zhanjiang, China
3
Methods: Adult zebrafish were fed a normal diet or a high-­fat diet combined with
School of Medical Technology,
Guangdong Medical University, egg yolk powder for 30 days. Body mass index (BMI) was measured to determine
Dongguan, China overall obesity. Serum lipids were measured using triglyceride (TG) and total choles-
Correspondence terol (TC) kits. Liver lipid deposition was detected by Oil Red O staining. Liver injury
Jingjing Zhang, Zhanjiang Key Laboratory was assessed by measuring glutathione aminotransferase (AST) and glutamic acid
of Zebrafish Model for Development and
Disease, Affiliated Hospital of Guangdong aminotransferase (ALT) levels. Reactive oxygen species (ROS) and malondialdehyde
Medical University, Zhanjiang 524001, (MDA) were used to evaluate oxidative damage. The level of inflammation was as-
China.
Email: jingjing.zhang@live.com sessed by qRT-­PCR for pro-­inflammatory factors. H&E staining was used for patho-

Jiangbin Li, School of Medical Technology,


logical histology. Caspase-­3 immunofluorescence measured apoptosis. Physiological
Guangdong Medical University, disruption was assessed via RNA-­seq analysis of genes at the transcriptional level and
Dongguan, 523808, China.
Email: jiangbinli@gdmu.edu.cn
validated by qRT-­PCR.
Results: The high-­fat diet led to significant obesity in zebrafish, with elevated BMI, he-
Honghui Guo, Department of Nutrition,
School of Public Health, Guangdong patic TC, and TG. Severe lipid deposition in the liver was observed by ORO and H&E
Medical University, Zhanjiang 524023,
staining, accompanied by massive steatosis and ballooning. Serum AST and ALT levels
China.
Email: guohh1999@gdmu.edu.cn were elevated, and significant liver damage was observed. The antioxidant system in
the body was severely imbalanced. Hepatocytes showed massive apoptosis. RNA-­seq
Funding information
the Discipline Construction Project of results indicated that several physiological processes, including endoplasmic reticu-
Guangdong Medical University, Grant/
lum stress, and glucolipid metabolism, were disrupted.
Award Number: 4SG22258G; the Science
and Technology Planning Project of Conclusion: Additional feeding of egg yolk powder to adult zebrafish for 30 consecu-
Zhanjiang, China, Grant/Award Number:
tive days can mimic the pathology of human nonalcoholic fatty liver disease.
2022A01231

KEYWORDS
ER stress, high-­fat-­diet, metabolism, non-­alcoholic fatty liver disease, zebrafish

Xiang Li and Lei Zhou contributed equally to this work.

This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in
any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
© 2023 The Authors. Animal Models and Experimental Medicine published by John Wiley & Sons Australia, Ltd on behalf of The Chinese Association for
Laboratory Animal Sciences.

Anim Models Exp Med. 2023;00:1–10.  wileyonlinelibrary.com/journal/ame2 | 1


|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2 LI et al.

1 | I NTRO D U C TI O N it does not induce the same pattern of NAFLD pathogenesis as


humans.19 High-­f at diet-­f ed mice are widely used since they pres-
Non-­alcoholic fatty liver disease (NAFLD) has become the most ent similar histological features and metabolic properties to those
common liver disease worldwide over the past few decades due to observed in humans. 20-­2 2 However, monitoring of rodents fed a
1
the harmful effects of overweight and obesity on human health. high-­f at diet involves high costs and time requirements, whereas
The progression of NAFLD includes a series of different degrees of the use of zebrafish allows for the rapid construction of an eco-
liver injuries, from nonalcoholic fatty liver (NAFL) to non-­alcoholic nomical and effective model of NAFLD. An et al. induced lipid ac-
steatohepatitis (NASH). 2 NAFL has a good prognosis, while NASH cumulation and oxidative stress by feeding a high-­c holesterol diet
may progress to advanced liver fibrosis, cirrhosis and even hepa- to zebrafish larvae. 23 Sapp et al. showed that fructose induced
tocellular carcinoma (HCC). 3 By 2018, the number of NAFLD pa- endoplasmic reticulum (ER) stress and oxidative stress in zebraf-
tients in the world had reached 25% of the total population,4 and ish larvae and successfully established a model of nonalcoholic
the prevalence of NAFLD in China was even as high as 29.2%. 5 In fatty liver disease. 24 However, most zebrafish NAFLD models use
spite of this huge medical demand, there is still no specific drug larvae. Although the age profile of NAFLD is getting younger, the
for the treatment of NAFLD/NASH in the clinic because of the juvenile version of NAFLD simulated by zebrafish larvae cannot
complex pathogenesis. 6 At present, the scientific community has fully reflect the pathogenesis in adults. An et al. established a
proposed more widely accepted pathogenesis mechanisms includ- model of NAFLD using adult zebrafish fed a high cholesterol diet
ing the two-­hit theory and the multiple hit theory, but the com- for 8 weeks. 25 Although the model was successfully established,
plexity of NAFLD still cannot be fully summarized.7,8 Therefore, it required a long experimental time.
the establishment of a fast and effective animal model of NAFLD In this study, we used adult male zebrafish as a vertebrate
is of great significance for exploring the molecular mechanism of model. We established NAFLD by additional feeding of egg yolk
NAFLD. powder, and evaluated the relationship between diet-­induced lipid
At present, the pathogenesis of NAFLD has been explored metabolism disorders and related pathways in the zebrafish. The
mainly using rodent models in vivo and cell models in vitro.9 After study aimed to more fully understand the effects of high-­f at diet
years of development, rodent models have yielded good results. on basic physical characteristics, liver histology and lipid metab-
However, rodent models inevitably involve high feeding costs, olism, thereby extending our understanding of the pathogenesis
large individual differences, and long experimental times, and are of NAFLD.
not suitable for large-­s ample screening.10 Most of the cell models
in vitro are single cell cultures, but the pathogenesis of NAFLD
is closely related to multiple tissues and organs, and because cell 2 | M ATE R I A L S A N D M E TH O DS
models grow in artificial environment, their morphology or func-
tion shows different degrees of change compared with the in vivo 2.1 | Care and feeding of adult Zebrafish
environment, meaning they cannot fully reflect real physiological
and pathological changes. Embryos of the wild-­t ype zebrafish AB strain were collected and
Zebrafish (Danio rerio) were first used in the laboratory placed in embryonic water, and incubated at 28.5°C in a constant
30 years ago. Due to its many advantages such as low mainte- temperature incubator until 5 days post fertilization (dpf). Paramecium
nance cost, rapid reproduction and development, and ease of in- and prawns were fed in still water from 6 dpf, and cultured in the sys-
heritance control and gene editing, the zebrafish has attracted tem at 15 dpf until adulthood (3 months post fertilization, mpf). Adult
more and more attention as an in vivo model in many fields such male zebrafish were randomly divided into a normal diet group (ND)
as toxicology, developmental biology and drug screening.11 The and a high-­fat diet group (HFD), with 15 fish in each group. The ND
zebrafish has become a model animal for studying NAFLD in group was fed with 30 mg of artemia per adult fish per day, while the
recent years because of its metabolic consistency with humans HFD group was fed with 30 mg of artemia (20% fat, 60% protein and
and the highly conserved molecular program of liver develop- polyunsaturated fatty acids; Shangjia, Shandong, China) per adult fish
ment.12,13 At present, several models of nonalcoholic fatty liver per day and 70 mg of egg yolk powder (containing 55.8% fat, 34.2%
disease have been established in zebrafish, including genetic, protein, 17.2% saturated fatty acids; purchased from Yuanye Bio-­
diet-­induced, and chemo-­induced models. 3,14,15,16The disad- Technology, Shanghai, China) per adult fish per day (egg yolk powder
vantage of genetic models is that mutant genes rarely occur in thoroughly mixed with 2 L water until no precipitation was observed).
humans.17 In contrast, drug-­induced models show high animal The feeding lasted for 30 days. After feeding, the animals were fasted
mortality. Diet-­induced models are widely used in the study of for a day before being killed. Based on standard laboratory guide-
molecular mechanisms because they are most similar to human lines, all zebrafish were fed in an environment at 28.5°C with a peri-
NAFLD pathogenesis. The MCD diet is a common model of NASH odic rhythm of 14 h of light and 10 h of darkness. The zebrafish were
and can rapidly cause steatosis, necrosis, inflammation, and fibro- treated according to the regulations of Guangdong Province on the
sis.18 However, its drawback is that as a nutrient-­d eficient diet, Administration of Experimental Animals (2010).
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
LI et al. 3

2.2 | Body mass index (BMI) 2.7 | Quantitative real-­time PCR

At the time of randomization (day 0), adult zebrafish were anesthe- Fresh adult zebrafish liver tissue was used to extract total RNA using
tized with embryonic water containing 0.02% Tricaine. After drying RNAiso Plus (Takara Bio), and a total of 1000 ng of RNA was reverse
the body surface water, the body weight (mg) was measured using transcribed into cDNA using a reverse transcription kit (Yeasen
an analytical balance, and the length (cm) from the fish mouth to Biotech). The cDNA obtained was mixed with specific primer pairs,
the end of the tail fin was measured with a ruler. The BMI of adult the Hieff® qPCR SYBR Green Master Mix (Yeasen Biotech) with Low
zebrafish was calculated at the beginning and end of the feeding ex- Rox and then subjected to real-­time PCR using the ABI QuantStudio
periment using the formula: BMI = weight/length2. 6 Flex system (Applied Biosystems). The sequences of primers are
listed in Table S1.

2.3 | Liver to body ratio (LBR)


2.8 | Immunofluorescence (IF)
Adult zebrafish were anesthetized and the abdomen was opened
with scissors and forceps to remove the intact liver. Livers were Samples were sectioned to 8 μm thick using a freezing microtome,
weighed using the analytical balance and the LBR was calculated fixed with acetone for 10 min on ice, and then washed with PBS.
using the formula: LBR = liver weight/body weight *100%. The samples were incubated with 5% goat serum for 1 h at room
temperature, and incubated with primary antibody (1:150, ab13847,
Abcam, UK) diluted with 5% goat serum overnight at 4°C in a re-
2.4 | Histology frigerator. The next day, the residual primary antibody was washed
with PBS. The sections were incubated with diluted secondary an-
The whole visceral mass of adult zebrafish sacrificed after anesthe- tibody (1:200, 111-­605-­0 03, Jackson, PA, USA) and DAPI (1:500,
sia was completely removed with scissors and forceps, embedded Sigma, USA) for 2 h at room temperature, washed with PBS, sealed
in Tissue-­Tek O.C.T. compound (SAKURA, USA), and frozen on dry with anti-­fluorescence quenched sealing (Beyotime Biotechnology,
ice. The samples were fixed on a Leica CM1860 UV cryostat for col- Shanghai, China) solution, and fixed with nail polish. Images were
lection of 8 μm thick serial sections on adhesive slides. After sam- obtained with an Olympus FV3000 confocal microscope.
ple collection, the sections were fixed in frozen methanol for 5 min.
After rinsing off excess methanol and O.C.T., the sections were
stained with H&E staining kit (Solarbio, Beijing, China) according 2.9 | RNA-­seq
to the manufacturer's instructions. Samples for ORO staining were
stained with 0.5% Oil Red O dye (Aladdin, Shanghai, China) solution The quality of RNA was examined using a Thermo Scientific
without methanol fixation. The staining results were analyzed under NanoPhotometer Spectrophotometer, Qubit2.0 Fluorameter and
a BX53 microscope (Olympus, Germany). an Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara,
CA, USA). High quality RNA extracts from each tissue sample were
pooled together for cDNA synthesis and sequencing. Fastp was used
2.5 | Biochemical analysis for quality control and the RNA-­seq reads were filtered to remove
low quality sequences. The processed reads were then mapped to a
After adult fish were sacrificed, intact livers were removed with reference zebrafish genome using HISAT2. The mRNAs and genes
scissors and forceps, and liver total cholesterol (TC), triglycerides with differential expression levels were screened out on the basis
(TG), reactive oxygen species (ROS), and malondialdehyde (MDA) of log2(fold change) >1 or < −1 and at p < 0.05 level. The subsequent
were measured using relevant kits according to the manufacturer's functional and signaling pathway analyses of the above-­screened
specifications. After centrifugation of blood taken from the tail of differential genes were conducted using Gene Ontology (GO; http://
the fish, serum alanine aminotransferase (ALT) and aspartate ami- curre​nt.geneo​ntolo​g y.org/) and Gene Set Enrichment Analysis
notransferase (AST) levels were measured using the appropriate kit (GSEA; http://www.gsea-­msigdb.org/).
(Jiancheng, Nanjing, China). 26-­28

2.10 | Statistical analyses


2.6 | Imaging
All data were statistically analyzed using Prism software (version 8.0
In order to obtain the liver changes in adult zebrafish, the abdomen GraphPad, San Diego, CA). The normality of data distribution was
was exposed and imaged using a Leica M205 FA stereo microscope analyzed using the Shapiro–­Wilk test. The unpaired Student's t test
(Leica, Germany). was used to compare the differences between groups. Data were
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4 LI et al.

expressed in the form of mean ± standard error of mean. p < 0.05 ND group (Figure 1B, left). After exposing the zebrafish belly, the
was considered as statistically significant. livers of the HFD group (Figure 1C, right) presented a more obvious
yellow color than those of ND zebrafish (Figure 1C, left). The results
of H&E staining showed that the livers of zebrafish in the HFD group
3 | R E S U LT S had inflammatory lesions (black arrow: aggregation of large numbers
of nuclei), ballooning degeneration (with blank regions in cytoplasm,
3.1 | Establishment of NAFLD zebrafish model or nucleus hanging in the center or squeezed to the side), and larger
areas of steatosis (p < 0.0001), indicating the progression of NAFLD
In order to establish the zebrafish model of NAFLD, adult male (Figure 1D). In the ND group fed with artemia at 50 mg/fish per day,
zebrafish were fed with artemia mixed with or without egg yolk the steatosis area reached 30%–­45% of total liver area. In the HFD
powder for 30 days (Figure 1A), and NAFLD was characterized by group fed 50 mg artemia/fish and 150 mg/fish egg yolk powder per
observing the accumulation of lipids in the liver. 29 At the end of the day, the steatosis area reached 35%–­50% (Figure S1). Based on the
feeding assay, the obesity phenotype of zebrafish in HFD group comparison of area of steatosis in the HFD group with that of the
(Figure 1B, right) was investigated and was compared with that in ND group (Figure 1D), the following experiments were carried out.

F I G U R E 1 Feeding protocol, zebrafish


body size and liver phenotype. (A) Feeding
protocol. Adult zebrafish (3 mpf) were
randomly divided into two groups. The
normal diet (ND) group was fed 30 mg
artemia/fish/day, while the high-­fat diet
(HFD) group was fed 30 mg artemia/
fish/day and 70 mg egg yolk/fish/day
powder. feeding lasted for 30 days. (B)
After completion of feeding, the HFD
group (right) showed a significantly obese
body shape compared to the ND group
(left). (C) After exposing the abdomen, the
liver of zebrafish in the HFD group (right)
appears distinctly yellow, while the liver of
zebrafish in the ND group (left) is orange-­
red. Scale bar: 1 mm. (D) H&E staining
results demonstrating steatosis and
inflammatory cell infiltration. In the ND
group 10–­20% steatosis was observed,
while in the HFD group 20–­30% steatosis
with marked inflammatory cell infiltration
was observed. The black arrow indicates
the regions where large numbers of
nuclei are aggregated and the ballooning
degeneration is seen as blank regions in
the cytoplasm with the nucleus hanging
in the center or squeezed to the side. The
solid black box indicates the magnified
area, and the high magnification image is
shown below. Scale bars: yellow, 100 μm;
black, 20 μm. 5 fields of view per fish (ND:
n = 14; HFD: n = 11). The rate of steatosis
was counted by Image J. Bar graphs,
mean ± SEM. ****p < 0.0001.
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
LI et al. 5

3.2 | HFD diet causes abnormal lipid accumulation 3.3 | HFD diet induced pathological changes in
in adult zebrafish adult zebrafish liver

At the beginning of the feeding experiment, there was no significant The serum levels of AST (Figure 3A) and ALT (Figure 3B) in the
difference in BMI between the two groups. At the end of the feed- HFD group (300.90 ± 37.39% and 180.10 ± 11.05%, respectively)
2
ing experiment, the BMI of the HFD group (25.60 ± 0.38 mg/cm ) were significantly higher than those in ND group (100.00 ± 5.00%
was significantly higher than that of the ND group (23.12 ± 0.33 mg/ and 100.00 ± 13.11%, respectively, p < 0.01). In addition, we meas-
cm2, p < 0.001, Figure 2A), indicating that significant obesity oc- ured liver ROS levels, which were significantly higher in the HFD
curred in the fish of the HFD group. LBR was also significantly in- group (6.52 ± 0.28) than in the control group (0.94 ± 0.18, p < 0.0001)
creased in the HFD group (1.90 ± 0.08%) compared to the ND group (Figure 3C). At the same time, we detected the level of MDA, the
(1.56 ± 0.08%, p < 0.01, Figure 2B), suggesting lipid accumulation in damaging oxidative product of lipid metabolism, and also found
the liver. The results of Oil Red O staining showed that a large num- that the level of the ND group (1.91 ± 0.79 mmol/mg protein) was
ber of red lipid droplets were present in the hepatocytes of the HFD significantly lower than that of the HFD group (6.91 ± 1.10 mmol/
group (13.12 ± 1.89%), while only a small number of lipid droplets mg protein, p < 0.01) (Figure 3D). qRT-­PCR results showed that the
were present in the ND group (0.57 ± 0.12%, p < 0.0001, Figure 2C). expression levels of proinflammatory factors (tnf-­α, il-­1β, il-­6, il-­8) in
Hepatic TC and TG levels were significantly lower in the ND group the HFD group were significantly higher than those in the ND group
(0.06 ± 0.01 mmol/g protein and 0.18 ± 0.02 mmol/g protein, re- (p < 0.01, p < 0.05 and p < 0.05, respectively) (Figure 3E). Caspase-­330
spectively) compared with the HFD group (0.16 ± 0.01 mmol/g pro- immunofluorescent staining results showed that severe apoptosis
tein and 0.26 ± 0.02 mmol/g protein, respectively, p < 0.0001 and occurred in the liver of the HFD group, while the level of apoptosis
p < 0.05, respectively; Figure 2D,E). These results indicate that the was lower in the ND group (Figure 3F). These results suggest that
HFD diet can lead to abnormal lipid accumulation in adult zebrafish. HFD diet can lead to pathological changes in adult zebrafish liver.

F I G U R E 2 High-­fat diet causes lipid


accumulation in zebrafish. (A) BMI at the
beginning and at the end of feeding. No
significant difference in BMI between
ND and HFD groups at the beginning of
feeding experiment was recorded (ND:
n = 14; HFD: n = 15), but at the end of
feeding, BMI was significantly higher in
the HFD group compared to the ND group
(ND: n = 14; HFD: n = 8). (B) Liver-­to-­
body ratio (LBR) was significantly higher
in the HFD group (ND: n = 13; HFD:
n = 8). (C) ORO staining showed large
lipid accumulation in the livers of the HFD
group. The solid black box indicates the
magnified area, and the high magnification
image is shown below. Scale bars: yellow
100 μm, black 20 μm. 5 fields of view per
fish (ND: n = 8; HFD: n = 6). The ratio of
red lipid droplets was counted by Image
J. (D) Zebrafish liver TC content (two
livers were combined into one sample,
ND: n = 4; HFD: n = 5). (E) Zebrafish liver
TG content (two livers pooled into one
sample, ND: n = 5; HFD: n = 5). Bar graphs
show mean ± SEM. n.s., not statistically
significant, *p < 0.05, **p < 0.01,
***p < 0.001, ****p < 0.0001.
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6 LI et al.

F I G U R E 3 High-­fat diet causes pathological changes in the liver of zebrafish. (A) Zebrafish serum AST levels (serum from 5 fish in one
sample, ND: n = 3; HFD: n = 4 repeats). (B) Zebrafish serum ALT levels (serum from 5 fish in one sample, ND: n = 3; HFD: n = 3 repeats).
(C) Zebrafish liver ROS levels (two livers pooled into one sample, ND: n = 4; HFD: n = 4 repeats). (D) Zebrafish liver MDA levels (two livers
pooled one sample, ND: n = 5; HFD: n = 5 repeats) (E) Liver RNA was extracted for qRT-­PCR assay to detect the expression levels of mRNAs
of inflammatory factors. (F) Hepatocyte apoptosis was observed by staining with Caspase-­3 protein. The expression level of Caspase-­3
was significantly increased in the HFD group compared with the ND group, and hepatocytes were extensively apoptotic. The solid white
box indicates the magnified area, and the high magnification image is shown below. Scale bars: white 100 μm, green 30 μm. 3 fields of view
per fish (ND: n = 4; HFD: n = 4 repeats). Mean grayscale values were counted by Image J. Bar graphs show mean ± SEM. n.s. means not
statistically significant, *p < 0.05, **p < 0.01, ****p < 0.0001.

3.4 | RNA-­seq analysis of changes in gene published results. 20,31,32,33 The genes associated with NAFLD in
expression levels during NAFLD progression the GO analysis were networked and enriched with the STRING
database, and then the cytoHubba app in Cytoscape software
Following a high-­f at diet, a volcano map showed that the expres- was used for module screening, and 20 core genes were ob-
sion of 293 genes was significantly increased and 452 genes were tained in order of score (Figure 4C). The expression of ER stress
significantly decreased in the high-­f at group (Figure S2). The re- molecules (hspa5, dnajc3), fatty acid synthase (fasn) and insulin
sults of GSEA and GO analysis showed that the expression of substrate receptor 2 (irs2) were significantly elevated in the HFD
genes related to the response to endoplasmic reticulum stress group as verified by qRT-­P CR (Figure 4D), demonstrating the oc-
and genes related to glycolipid metabolism were upregulated in currence of ER stress and disorders of glucolipid metabolism in
the HFD group (Figure 4A,B). This is consistent with previously the model group.
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
LI et al. 7

F I G U R E 4 RNA-­seq analysis of changes in gene transcript levels during NALFD progression. (A) GSEA analysis of differentially expressed
genes with significant upregulation of response pathways to endoplasmic reticulum stress. (B) Bubble plot of GO analysis restlt, lipid
synthesis, insulin response and endoplasmic reticulum stress pathways were significantly upregulated. (C) Screening results of cytoHubba
app module with the top 20 highest scoring genes. (D) qRT-­PCR validation of mRNA expression levels of endoplasmic reticulum stress
molecules, fatty acid synthase and insulin substrate receptor (more than three samples were tested for each gene). n.s., no statistical
significance, *p < 0.05, **p < 0.01, ***p < 0.001.

4 | DISCUSSION the water to reduce any differences in feeding. Second, to obtain


suitable feeding concentrations, we performed feeding experiments
In this study, a model for studying NAFLD was constructed using using two concentrations and compared them by H&E staining. It
egg yolk powder fed to adult zebrafish; this model is more consist- was found that the ND group fed 30 mg/fish/day had a lower area
ent with human epidemiological characteristics than models using of steatosis and did not produce inflammatory lesions, which better
juvenile fish. In addition, additional intake of high-­fat diet beyond mimicked conditions in the normal physiological state. Therefore,
that required to meet basic physiological needs can better mimic 30 mg artemia /fish per day was chosen as the basal feeding level in
the pathogenesis and pathological characteristics of human NAFLD. subsequent experiments.
Importantly, the dose of egg yolk powder used in the study allowed Under normal conditions, only a very small amount of TG is
the model to be completed in a shorter period of time. stored in the lipid droplets of hepatocytes in the liver. However, ex-
First, to ensure that each zebrafish consumed the same amount cessive storage of TG leads to the development of lipotoxicity and
of food, we selected zebrafish of similar body size as the experi- steatosis.34,35 TC in vivo includes cholesteryl esters and free choles-
mental subjects, and we dispersed the egg yolk powder evenly in terol (FC), and the accumulation of FC has been shown to correlate
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
8 LI et al.

with inflammation and fibrosis, among other symptoms, in the pro- receptor substrate 2 (IRS2) regulates the AKT pathway and affects
36
gression of NASH. Elevated TG and TC were detected in our ex- NAFLD progression.47 Consistent with the results of Chi et al,48
perimental results, and the results of Oil Red O staining of frozen we detected increased mRNA expression of irs2. Finally, the en-
sections also showed the presence of a large number of lipid drop- doplasmic reticulum plays an important role in lipid biosynthesis
lets in the HFD group, confirming the excessive accumulation of TG. in vivo,49 and consistent with previous findings, 50,51 we detected
In addition, the results of H&E staining also showed a large extent of increased expression of the ER stress chaperone molecule heat
fatty degeneration and inflammatory lesions. shock protein 5 (hspa5) and the DNAJ heat shock protein family
An imbalance between the bioavailability of ROS and the antiox- member C3 (danjc3) in the HFD group. In conclusion, a high-­f at diet
idant system in the body can lead to the development of oxidative induces upregulation of the DNL pathway and increased lipogene-
stress. Low levels of ROS play physiological roles such as signaling in sis, leading to excessive accumulation of lipid droplets in the liver.
vivo, but high levels of ROS can lead to DNA damage, which in turn Lipid accumulation produces lipotoxicity, leading to mitochondrial
can cause alterations in the expression of certain genes, ultimately dysfunction or endoplasmic reticulum stress, which, together with
leading to increased cell death. We detected a significant increase in oxidative stress, activation of pro-­inflammatory substances and
ROS levels in the HFD group, indicating that the antioxidant balance apoptotic pathways, leads to hepatocyte death and promotes the
was disrupted. In addition, high levels of ROS were able to induce development of NAFLD.
protein and lipid peroxidation, leading to functional alterations of Currently, research on the pathogenesis of NAFLD is limited,
substances such as enzymes and cell membranes. We detected a and no drugs have been approved for treatment.14 Previously pro-
significant increase in MDA levels in the HFD group. Excessive accu- posed theories such as the ‘double-­hit’ theory based on triglycer-
mulation of ROS affects the mitochondrial depolarization potential ide accumulation, inflammation, oxidative stress and autophagy,8
by inducing lipid peroxidation reactions to produce MDA. In the mi- and the ‘three strike’ theory52 based on steatosis, lipotoxicity and
tochondrial depolarized state, the burden of mitochondrial autoph- inflammation, have failed to summarize the complexity of human
agy increases and the number of mitochondria decreases, promoting NAFLD. In recent years, the ‘multiple-­hit’ theory, which focuses on
further development of steatosis, inflammation and fibrosis.37-­39 mitochondrial dysfunction, endoplasmic reticulum stress, inflamma-
Hepatocyte injury usually releases AST and ALT into the blood. tory response, lipotoxicity, genetic factors, nutritional excess and
Therefore, biochemical assays of serum AST and ALT are widely intestinal flora disorders, has flourished.7 Here, we constructed a
used as markers of liver injury.40 In zebrafish in the HFD group, sig- short, convenient, and high-­volume experimental model of NAFLD
nificantly elevated serum AST and ALT levels were detected, sug- in adult zebrafish. 3mpf male zebrafish were additionally fed with
gesting that the high-­fat diet induced the process of liver injury. egg yolk powder for 30 days, and relevant indexes were tested to
In recent years, as the pathogenesis of NAFLD/NASH has been verify the success of the model, which successfully mimicked the
increasingly studied, the role played by endoplasmic reticulum pathological characteristics of NAFLD and will hopefully provide a
(ER) stress has received wide attention and ER stress has been stable and reliable in vivo model for drug screening and the study of
shown to play a regulatory role in apoptosis of hepatocytes.41 In NAFLD pathogenesis.
the presence of ER stress, the unfolded protein response (UPR)
is initiated and undergoes repair. However, prolonged ER stress AU T H O R C O N T R I B U T I O N S
leads to a shift from adaptive to terminal UPR, followed by in- Xiang Li: carried out the animal experiment and drafted the manu-
creased inflammation and activation of pro-­a poptotic pathways script. Yuying Zheng: information collection and revised the manu-
in the liver, resulting in massive hepatocyte death.42,43 Using a script. LeiZhou: design methodology and revision of manuscript.
immunofluorescence assay, we detected a large amount of the Honghui Guo, Jiangbin Li and Taiping He: supervision, extra sugges-
apoptotic marker Caspase-­3 in the HFD group, suggesting that tions on this study. Jingjing Zhang: supervision, conceptualization,
ER stress may have occurred in zebrafish in the HFD group. The writing -­review and editing.
dynamic mechanisms regulating ER stress in NAFLD are not fully
understood, and although extensive studies have been conducted AC K N OW L E D G M E N T S
and many relevant molecules have emerged as new therapeutic We thank Yuezhuang Zheng, Qiumei Hong and Weinan Wu for ex-
targets, more in-­d epth experiments are needed to explore the pert technical assistance with the zebrafish facility. This work was
complete mechanisms.44 supported by thePublic ServicePlatform of South China Sea for the
Analysis of the results of RNA-­seq and qRT-­P CR assays al- R&D of Marine BiomedicineResource for support.
lowed us to identify several biochemical processes that are most
altered in this model. First, fatty acid synthase (FASN) is a key F U N D I N G I N FO R M AT I O N
rate-­limiting enzyme for hepatic de novo lipogenesis (DNL),45,46 This study was supported by the Science and Technology
and we found that a high-­f at diet induced a rise in FASN expres- Planning Project of Zhanjiang, China (2022A01231) and the
sion, leading to increased lipid synthesis and further leading to Discipline Construction Project of Guangdong Medical University
TG accumulation in the liver. Second, altered expression of insulin (4SG22258G).
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
LI et al. 9

method for non-­alcoholic fatty liver disease drugs. Int J Biol Sci.
C O N F L I C T O F I N T E R E S T S TAT E M E N T 2019;155:973-­983.
The authors declare that they have no conflicts of interest. 15. Sun L, Ling Y, Jiang J, et al. Differential mechanisms regarding triclosan
Jingjing Zhang is an editorial board member of Animal Models and vs. bisphenol a and fluorene-­9-­bisphenol induced zebrafish lipid-­
metabolism disorders by RNA-­seq. Chemosphere. 2020;251:126318.
Experimental Medicine and a coauthors of this article. To minimize
16. Templehof H, Moshe N, Avraham-­Davidi I, et al. Zebrafish mutants
bias, he was excluded from all editorial decision making related to
provide insights into apolipoprotein B functions during embryonic de-
the acceptance of this article for publication. velopment and pathological conditions. JCI Insight. 2021;6:e130399.
17. Kanuri G, Bergheim I. In vitro and in vivo models of non-­alcoholic
E T H I C S S TAT E M E N T fatty liver disease (NAFLD). Int J Mol Sci. 2013;146:11963-­11980.
18. Li X, Wang T-­X , Huang X, et al. Targeting ferroptosis alleviates
This study does not contain any studies with human subjects or
methionine-­choline deficient (MCD)-­diet induced NASH by sup-
with rodent vertebrates. Handling of zebrafish was performed in ac- pressing liver lipotoxicity. Liver Int. 2020;406:1378-­1394.
cordance with Guangdong State Regulations on Laboratory Animal 19. Zhong F, Zhou X, Xu J, Gao L. Rodent models of nonalcoholic fatty
Management (2010). The whole experimental procedures were ex- liver disease. Digestion. 2020;1015:522-­535.
20. Smith GI, Shankaran M, Yoshino M, et al. Insulin resistance drives
amined by the Clinical Ethics Committee of the Affiliated Hospital of
hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J Clin
Guangdong Medical University, China. Invest. 2020;1303:1453-­1460.
21. Wang W, Zhao J, Gui W, et al. Tauroursodeoxycholic acid inhibits
ORCID intestinal inflammation and barrier disruption in mice with non-­
alcoholic fatty liver disease. Br J Pharmacol. 2018;1753:469-­484.
Xiang Li https://orcid.org/0000-0002-8193-7342
22. Zhou T, Chang L, Luo Y, Zhou Y, Zhang J. Mst1 inhibition attenuates
Honghui Guo https://orcid.org/0000-0001-7837-0392 non-­alcoholic fatty liver disease via reversing parkin-­related mito-
Jingjing Zhang https://orcid.org/0000-0002-8789-4638 phagy. Redox Biol. 2019;21:101120.
23. An J, Cheng L, Yang L, et al. P-­ hydroxybenzyl alcohol alleviates
oxidative stress in a nonalcoholic fatty liver disease larval zebrafish
REFERENCES
model and a BRL-­3A hepatocyte the Nrf2 pathway. Front Pharmacol.
1. Eslam M, Valenti L, Romeo S. Genetics and epigenetics of NAFLD 2021;12:646239.
and NASH: clinical impact. J Hepatol. 2018;682:268-­279. 24. Sapp V, Gaffney L, EauClaire SF, et al. Fructose leads to hepatic
2. Friedman SL, Neuschwander-­Tetri BA, Rinella M, et al. Mechanisms steatosis in zebrafish that is reversed by mechanistic target of rapa-
of NAFLD development and therapeutic strategies. Nat Med. mycin (mTOR) inhibition. Hepatology. 2014;605:1581-­1592.
2018;247:908-­922. 25. An HJ, Lee YJ, Choe CP, et al. Long noncoding RNAs associated
3. Wang C, Hu N-­H, Yu L-­Y, et al. 2,3,5,4′-­tetrahydroxystilbence-­2-­O with nonalcoholic fatty liver disease in a high cholesterol diet adult
-­β-­D-­glucoside attenuates hepatic steatosis via IKKβ/NF-­κB and zebrafish model. Sci Rep. 2021;111:23005.
Keap1-­Nrf2 pathways in larval zebrafish. Biomed Pharmacother. 26. Zou Y, Chen Z, Sun C, et al. Exercise intervention mitigates patho-
2020;127:110138. logical liver changes in NAFLD zebrafish by activating SIRT1/
4. Younossi Z, Tacke F, Arrese M, et al. Global perspectives on non- AMPK/NRF2 signaling. Int J Mol Sci. 2021;22:10940.
alcoholic fatty liver disease and nonalcoholic steatohepatitis. 27. Zhu Z, Zhang Y, Huang X, et al. Thymosin beta 4 alleviates non-­
Hepatology. 2019;696:2672-­2682. alcoholic fatty liver by inhibiting ferroptosis via up-­regulation of
5. Zhou J, Zhou F, Wang W, et al. Epidemiological features of NAFLD GPX4. Eur J Pharmacol. 2021;908:174351.
from 1999 to 2018 in China. Hepatology. 2020;715:1851-­1864. 28. Yu Q, Huo J, Zhang Y, et al. Tamoxifen-­induced hepatotoxicity via
6. Sodum N, Kumar G, Bojja SL, Kumar N, Rao CM. Epigenetics in NAFLD/ lipid accumulation and inflammation in zebrafish. Chemosphere.
NASH: targets and therapy. Pharmacol Res. 2021;167:105484. 2020;239:124705.
7. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-­hit pathogen- 29. Ferguson D, Finck BN. Emerging therapeutic approaches for
esis of non-­alcoholic fatty liver disease (NAFLD). Metabolism. the treatment of NAFLD and type 2 diabetes mellitus. Nat Rev
2016;658:1038-­1048. Endocrinol. 2021;178:484-­495.
8. Engin A. Non-­alcoholic fatty liver disease. Adv Exp Med Biol. 30. Wang W, Zhu M, Xu Z, et al. Ropivacaine promotes apoptosis of
2017;960:443-­467. hepatocellular carcinoma cells through damaging mitochondria and
9. Willebrords J, Pereira IVA, Maes M, et al. Strategies, models and activating caspase-­3 activity. Biol Res. 2019;521:36.
biomarkers in experimental non-­alcoholic fatty liver disease re- 31. Kim JY, Garcia-­C arbonell R, Yamachika S, et al. ER stress drives
search. Prog Lipid Res. 2015;59:106-­125. lipogenesis and steatohepatitis via Caspase-­2 activation of S1P.
10. Chen B, Zheng Y-­M, Zhang J-­P. Comparative study of differ- Cell. 2018;175:133-­145.
ent diets-­induced NAFLD models of zebrafish. Front Endocrinol 32. Zhang L, Zhang Z, Li C, et al. S100A11 promotes liver steatosis via
(Lausanne). 2018;9:366. FOXO1-­mediated autophagy and lipogenesis. Cell Mol Gastroenterol
11. García-­Moreno D, Tyrkalska SD, Valera-­Pérez A, Gómez-­Abenza Hepatol. 2021;113:697-­724.
E, Pérez-­Oliva AB, Mulero V. The zebrafish: a research model to 33. Long J-­K , Dai W, Zheng Y-­W, et al. miR-­122 promotes hepatic lipo-
understand the evolution of vertebrate immunity. Fish Shellfish genesis via inhibiting the LKB1/AMPK pathway by targeting Sirt1 in
Immunol. 2019;90:215-­222. non-­alcoholic fatty liver disease. Mol Med. 2019;251:26.
12. Wilkins BJ, Pack M. Zebrafish models of human liver development 34. Kawano Y, Cohen DE. Mechanisms of hepatic triglyceride ac-
and disease. Compr Physiol. 2013;33:1213-­1230. cumulation in non-­alcoholic fatty liver disease. J Gastroenterol.
13. Winata CL, Dodzian J, Bialek-­Wyrzykowska U. The zebrafish as a 2013;484:434-­4 41.
model for developmental and biomedical research in Poland and 35. Alves-­Bezerra M, Cohen DE. Triglyceride metabolism in the liver.
beyond. Dev Biol. 2020;4572:167-­168. Compr Physiol. 2017;81:1-­8.
14. Ma J, Yin H, Li M, et al. A comprehensive study of high choles- 36. Marra F, Svegliati-­Baroni G. Lipotoxicity and the gut-­liver axis in
terol diet-­induced larval zebrafish model: a short-­time screening NASH pathogenesis. J Hepatol. 2018;682:280-­295.
|

25762095, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ame2.12309 by UECE - Universidade Estadual do Ceara, Wiley Online Library on [23/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10 LI et al.

37. Farzanegi P, Dana A, Ebrahimpoor Z, et al. Mechanisms of benefi- 47. Jeong S-­H, Kim H-­B, Kim M-­C , et al. Hippo-­mediated suppression
cial effects of exercise training on non-­alcoholic fatty liver disease of IRS2/AKT signaling prevents hepatic steatosis and liver cancer.
(NAFLD): roles of oxidative stress and inflammation. Eur J Sport Sci. J Clin Invest. 2018;1283:1010-­1025.
2019;19:994-­1003. 48. Chi Y, Gong Z, Xin H, et al. Long noncoding RNA lncARSR promotes
38. Rives C, Fougerat A, Ellero-­Simatos S, et al. Oxidative stress in nonalcoholic fatty liver disease and hepatocellular carcinoma by
NAFLD: role of nutrients and food contaminants. Biomolecules. promoting YAP1 and activating the IRS2/AKT pathway. J Transl
2020;10:1702. Med. 2020;181:126.
39. Paradies G, Paradies V, Ruggiero FM, et al. Oxidative stress, car- 49. Stevenson J, Huang EY, Olzmann JA. Endoplasmic reticulum-­
diolipin and mitochondrial dysfunction in nonalcoholic fatty liver associated degradation and lipid homeostasis. Annu Rev Nutr.
disease. World J Gastroenterol. 2014;2039:14205-­14218. 2016;36:511-­542.
40. Xiong X, Ren Y, Cui Y, Li R, Wang C, Zhang Y. Obeticholic acid pro- 50. Jin M, Wei Y, Yu H, et al. Erythritol improves nonalcoholic fatty liver
tects mice against lipopolysaccharide-­induced liver injury and in- disease by activating Nrf2 antioxidant capacity. J Agric Food Chem.
flammation. Biomed Pharmacother. 2017;96:1292-­1298. 2021;6944:13080-­13092.
41. Oakes SA, Papa FR. The role of endoplasmic reticulum stress in 51. Vig S, Buitinga M, Rondas D, et al. Cytokine-­induced translocation
human pathology. Annu Rev Pathol. 2015;10:173-­194. of GRP78 to the plasma membrane triggers a pro-­apoptotic feed-
42. Nasiri-­Ansari N, Nikolopoulou C, Papoutsi K, et al. Empagliflozin back loop in pancreatic beta cells. Cell Death Dis. 2019;104:309.
attenuates non-­alcoholic fatty liver disease (NAFLD) in high fat diet 52. Cobbina E, Akhlaghi F. Non-­alcoholic fatty liver disease (NAFLD) –­
fed ApoE mice by activating autophagy and reducing ER stress and pathogenesis, classification, and effect on drug metabolizing en-
apoptosis. Int J Mol Sci. 2021;22:818. zymes and transporters. Drug Metab Rev. 2017;492:197-­211.
43. Lebeaupin C, Vallée D, Hazari Y, Hetz C, Chevet E, Bailly-­M aitre
B. Endoplasmic reticulum stress signalling and the pathogen-
esis of non-­a lcoholic fatty liver disease. J Hepatol. 2018;694:​
927-­947. S U P P O R T I N G I N FO R M AT I O N
44. Xu H, Tian Y, Tang D, et al. An endoplasmic reticulum stress-­ Additional supporting information can be found online in the
MicroRNA-­26a feedback circuit in NAFLD. Hepatology. 2021;734:​ Supporting Information section at the end of this article.
1327-­1345.
45. Du X, Cai C, Yao J, et al. Histone modifications in FASN mod-
ulated by sterol regulatory element-­b inding protein 1c and car-
bohydrate responsive-­e lement binding protein under insulin How to cite this article: Li X, Zhou L, Zheng Y, et al.
stimulation are related to NAFLD. Biochem Biophys Res Commun. Establishment of a non-­alcoholic fatty liver disease model by
2017;4831:409-­417.
high fat diet in adult zebrafish. Anim Models Exp Med.
46. Hu Y, He W, Huang Y, et al. Fatty acid synthase-­suppressor screen-
ing identifies sorting nexin 8 as a therapeutic target for NAFLD. 2023;00:1-10. doi:10.1002/ame2.12309
Hepatology. 2021;745:2508-­2525.

You might also like