You are on page 1of 13

Food &

Function
View Article Online
PAPER View Journal

Alginate oligosaccharide (AOS) improves


immuno-metabolic systems by inhibiting
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

Cite this: DOI: 10.1039/c9fo00982e


STOML2 overexpression in high-fat-diet-
induced obese zebrafish†
a,b c
Van Cuong Tran, Se-Young Cho, Joseph Kwon*c and Duwoon Kim *a,d

This study aimed to evaluate the potentially beneficial effects of alginate oligosaccharide (AOS) on the
modulation of immuno-metabolic pathways in high-fat-diet (HFD)-induced obese zebrafish and the
underlying mechanism. AOS showed a marked anti-obesity effect in that it reduced body weight, BMI,
and the blood glucose level. To understand the mechanisms of action of AOS, comparative proteomics
was performed through UPLC-HDMSE analysis between HFD vs. normal diet (NFD) and HFD + AOS vs.
HFD. Among 146 proteins differentially modulated by AOS in HFD-induced obesity zebrafish, STOML2
(Stomatin-like protein 2) was selected as a specific biomarker. AOS suppressed obesity and pathophysio-
Received 7th May 2019, logical disorders in HFD-fed zebrafish by modulating lipid metabolism, suppressing inflammation, down-
Accepted 2nd July 2019
regulating apoptosis-related genes, and improving immune function by inhibiting STOML2. Our results
DOI: 10.1039/c9fo00982e suggest that STOML2 can serve as a platform for further studies to discover novel treatments for meta-
rsc.li/food-function bolic disorders. AOS might be useful as a dietary health supplement, especially for reducing obesity.

1. Introduction Zebrafish are a popular model for studying metabolic


disorders and infectious diseases, toxicological studies, and
Alginate oligosaccharide (AOS) is a depolymerization product drug screening.16–19 Zebrafish have been used as a model for
of sodium alginate that has attracted increasing attention high-fat-diet-induced obesity,20–22 as lipid metabolism in
in a wide variety of industries, including the food,1,2 zebrafish is very similar to that in humans.20 The zebrafish
agricultural,3–5 and pharmaceutical industries.6 Sodium algi- genome has been sequenced, and the zebrafish physiology is
nate and its derivatives confer health benefits owing to their very similar to that of mammals; therefore, in the present
diverse biological activities, such as antioxidant,7,8 antifungal,9 study, we used zebrafish to investigate how dietary AOS regu-
anti-apoptotic,10,11 anti-inflammatory,12 and anti-tumor lates immuno-metabolic pathways. We conducted a compara-
effects.13,14 AOS has shown anti-obesity effect in vitro and tive proteomics analysis of high-fat-diet (HFD)- and normal-
in vivo.15 Nevertheless, there are no reports on the impact of diet (NFD)-fed animals.
dietary AOS on the regulation of the immuno-metabolic path- STOML2 (Stomatin-like protein 2) is a mitochondrial
ways in zebrafish on a high-fat diet. protein that is considered a cancer-related gene.23,24 STOML2
is overexpressed in various cancers, including human
non-small cell lung cancer cells, laryngeal carcinoma,
breast cancer, and gastric cancer, cervical cancer.23,25–27
a
Department of Food Science and Technology, Chonnam National University, Mitochondrial dysfunction is known to be linked to obesity.28
Gwangju, 61186, Republic of Korea. E-mail: dwkim@jnu.ac.kr; Fax: +82 62 530 2149
STOML2 overexpression might contribute to metabolism and
; Tel: +82 62 530 2144
b
Department of Food Science and Post-harvest Technology, Tay Nguyen University,
inflammatory responses.29 Further, there is a strong corre-
Daklak, Vietnam lation between obesity and cancers.30 Therefore, STOML2 may
c
Biological Disaster Analysis Group, Korea Basic Science Institute, Daejeon, 34133, play a role in obesity development. However, its mechanism of
Republic of Korea. E-mail: joseph@kbsi.re.kr; Fax: +82 42 865 3419; action is not well characterized, and effects of STOML2 on
Tel: +82 42 865 3446
d
metabolic syndromes, especially obesity, have not been docu-
Foodborne Virus Research Center, Chonnam National University, Gwangju, 61186,
Republic of Korea
mented. As STOML2 is predicted to be a critical regulatory
† Electronic supplementary information (ESI) available. See DOI: 10.1039/ protein, we explored its involvement in various pathways
c9fo00982e involved in HFD-induced obesity in zebrafish.

This journal is © The Royal Society of Chemistry 2019 Food Funct.


View Article Online

Paper Food & Function

2. Materials and methods To determine blood glucose levels, three to four fish were
randomly collected from each group and fasted overnight
2.1. Preparation and characterization of alginate (∼12 h). Blood samples were collected from the individual zeb-
oligosaccharide (AOS) rafish at the dorsal artery and glucose was immediately
Sodium alginate (Wako, Osaka, Japan) was used to produce measured with a glucometer (CareSens-II Plus+; Informa Life
depolymerized alginate (AOS) by using bacterial alginate lyase. Sciences) twice for each zebrafish.
The sodium alginate in phosphate buffer ( pH 7.0) was mixed
with the lyase and incubated at 30 °C for 24 h. After centrifu- 2.4. Protein isolation and relative quantification
gation at 12 000g for 15 min, AOS in the supernatant was con- After the five-week experiment, five fish from each group were
centrated by ultrafiltration through a polysulfone membrane pooled, frozen in liquid nitrogen, and grounded to form a
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

(VivaFlow 50; Sartorius, AG, Germany) with a molecular weight powder. Proteins were extracted using Pierce RIPA buffer
(MW) cut-off of 3000 Da, and lyophilized. (Thermo Fisher Scientific, Waltham, MA, USA) containing
The MW of AOS was analyzed by gel-filtration chromato- 1.0% Halt Protease Inhibitor Cocktail 100× (Thermo Fisher
graphy. The samples were separated on Ultrahydrogel 120, 250, Scientific, Waltham, MA, USA), 1.0% Phosphatase Inhibitor
500, and 1000 (1.8 × 300 nm) columns using a high-perform- Cocktail (Sigma-Aldrich, St Louis, MO, USA), and 1.0% EDTA
ance size-exclusion chromatography system (e2695 Separations 0.5 M (Thermo Fisher Scientific, Waltham, MA, USA). The pro-
Module; Waters Corporation, Milford, MA, USA) with a built-in teins were digested by tube-gel digestion,31 and subjected to
refractive index detector, and 0.1 M sodium phosphate buffer nano-ultra-performance liquid chromatography (UPLC) and
(pH 7.0) was used as an eluent. Calibration curves were obtained Synapt G2-Si High Definition Mass Spectrometry (HDMSE,
from polyethylene glycol. The relative MW was calculated by Waters Corp., USA), as previously described.32 Proteins were
comparing the retention times of the standard and sample. identified and quantified using Progenesis QI for Proteomics
To determine the M/G ratio in AOS, circular dichroism version 2.0 (Nonlinear Dynamics, Newcastle, UK).33 Protein
experiments were carried out with a J-1500 spectrometer lists were first defined using the International Protein Index
(JASCO, Tokyo, Japan). The absorbance of AOS dissolved in database, and then re-identified using the UniProt database
phosphate-buffered solution (2.5 mg mL−1) was measured for zebrafish (http://www.uniprot.org).
from 190 to 260 nm at 25 °C using a 1 mm-path cell with the
following setup: bandwidth, 1 nm; time constant, 1 s; scan- 2.5. Bioinformatics and network analyses
ning speed, 200 nm min−1. Three spectra corrected for the After identification and relative quantification, only proteins
background were averaged for the sample. containing at least one unique peptide were considered valid
for identification. Identified proteins were hierarchically clus-
2.2. Zebrafish care and diets tered based on their expression (log base 2), and a heatmap of
Adult zebrafish (Danio rerio) with body weights of 200–300 mg significantly differentially expressed proteins was generated.
and approximately three months old were purchased from a K-means clustering was performed based on Euclidean dis-
local aquarium shop in Gwangju, Korea and were kept under a tance with average linkage, using Multi Experiment Viewer
14 h light/10 h dark cycle (lights on: 9 am, lights off: 11 pm) at version 4.9.0 (http://mev.tm4.org/).34 Ingenuity Pathway
28 ± 0.5 °C for a week. After acclimatization, 30 fish per group Analysis (IPA version 9.0; Ingenuity Systems Inc., http://www.
were randomly assigned to three experimental groups: normal ingenuity.com) was used to identify biological functions and
diet (NFD), HFD, and HFD supplemented with AOS (HAOS). canonical pathways of related proteins derived from proteo-
Fish in each group were maintained in a 4 L aquarium tank. mics data. Gene set enrichment analysis (GSEA) (http://software.
During the 5 week experiment, fish in all groups were fed broadinstitute.org/gsea/index.jsp) was performed to identify func-
twice daily (at 9:00 am and 5:00 pm; Fig. S2†). The NFD was a tional classes and biological pathways of target molecules.35
commercial feed (Tetra Bits Complete), and for the HFD, 25%
of lard was added to the commercial diet. For HAOS, 2.5% AOS 2.6. Inhibitor/activator treatments
was added to the HFD. All zebrafish experiments were con- To verify the roles of AOS and to clarify whether its effect on
ducted according to the Zebrafish Book (visited at https://zfin. immuno-metabolic pathways may be mediated by STOML2 in
org/zf_info/zfbook/zfbk.html) and in accordance with the insti- zebrafish, we used various inhibitors or activators, including
tutional animal care guidelines and with supervision of the AICAR, U0126, KNK437, and LPS (details are given in
relevant committees of Chonnam National University (Permit Table S1†). Zebrafish were given NFD or HFD for four weeks
number: CNU IACUC-YS20135). until obesity-related phenotypes were observed. Stock solu-
tions of the above compounds were prepared in DMSO (Sigma-
2.3. Body weight and fasting blood glucose measurements Aldrich, St Louis, MO, USA) at 1 mg mL−1 or 10 mg mL−1, per
During the five-week feeding period, five zebrafish were ran- the supplier’s recommendations. For administration to the
domly collected from each group on day 1 and then weekly to zebrafish, the stock solutions were diluted in phosphate-
determine the body weight. At the same time, the body length buffered solution. AICAR and AOS were administered at 100
(mm) was recorded and the BMI (mg mm−2) was calculated as mg kg−1 day−1, and U0126, KNK437, and LPS at 10 mg kg−1 day−1
described previously.20 once daily in 10 μL doses using a micro-pipette. Zebrafish were

Food Funct. This journal is © The Royal Society of Chemistry 2019


View Article Online

Food & Function Paper

treated for three days and then 3–4 zebrafish/treatment were nuronic acid obtained from sodium alginate reported peak at
sampled for further analysis. 200 nm and a trough at 215 nm.36 As shown in Fig. S1B,†
sodium alginate was composed of 58.02% mannuronate (M)
2.7. RNA isolation and cDNA transcription and 41.98% guluronate (G) as the M/G of 1.38 : 1.0, while the
Total RNA was extracted from the whole bodies of adult zebra- M/G ratio of AOS was calculated to be 1.51 : 1.0 based on mdeg
fish using RNAiso Plus reagent (Takara Bio, Shiga, Japan) values, and the depolymerized sodium alginate consisted of
according to the manufacturer’s instructions. cDNA was syn- 60.1% mannuronate and 39.9% guluronate. Thus, the product
thesized from 10 μg of RNA in a reaction mixture containing prepared from sodium alginate by bacterial alginate lyase was
1 μL random primer, 2 μL dNTPs, 0.5 μL recombinant RNase a mannuronate-rich alginate, which we termed “AOS”. The
inhibitor (Takara Bio), 1 μL MMLV RT, 4 μL 5× MMLV RT unsaturated AOS had a characteristic peak at 234 nm and
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

buffer, and 2 μL 5× DTT (Beams Biotechnology, Seongnam, maximal ellipticity at 250 nm, which can be explained by the
Korea), on a Takara PCR Thermal Cycler Dice Real Time properties of unsaturated alginate decomposed by alginate lyase.37
System under the following conditions: 63 °C for 10 min,
3.2. AOS suppresses HFD-induced obesity in zebrafish
37 °C for 60 min, and 95 °C for 5 min.
Previous studies have reported that HFD/high cholesterol diet
2.8. Quantitative real time (qRT-)PCR or overfeeding induce obesity in adult zebrafish.20–22,38
Genes related to lipid metabolism, endoplasmic reticulum Nakazono et al. reported that alginate oligomer exerted anti-
(ER) stress, stress-induced apoptosis and inflammation, and obesity effects in mice fed a HFD by lowering serum triglycer-
immune responses were targeted using gene-specific primers ide and pancreatic lipase and by inhibiting gastrointestinal
(details in Table S2†). qRT-PCR was conducted in a Thermal lipid absorption.15 To investigate the potentially beneficial
Cycler Dice Real Time System (Takara Bio) using 21 μL reaction effects of AOS in the obese zebrafish model, adult zebrafish
mixtures containing 10 μL SYBR Green Master Kit (Takara were fed a NFD, HFD, or HAOS for five weeks (Fig. S2†), and
Bio), 1 μL of each of forward and reverse primers, 2 μL cDNA, changes in body weight, BMI, and fasting blood glucose were
and 7 μL RNase-free water. Thermal cycles were as follows: 30 s measured. As expected, after the five-week experiment, the fish
hold at 95 °C, 45 cycles of 5 s at 95 °C, 10 s at 55 °C, and 20 s had significantly gained weight in all groups as compared to
at 72 °C, and a cycle of dissociation at 95 °C for 15 s, 60 °C for their initial weights, indicating normal growth. The HFD
30 s and 95 °C for 15 s. The β-actin gene was used for normali- group showed a significantly higher increase (1.22-fold or
zation. Target mRNA levels were determined by the 2–ΔΔCt 22.14%) in body weight than the NFD group (Fig. 1A–C). AOS
method. All qRT-PCRs were run in triplicate. attenuated weight gain; in the HAOS-group, mean body weight
was only 1.10-fold (10.68%) higher than in the NFD group
2.9. Statistical analysis after five weeks of feeding (Fig. 1C). Similarly, the BMI showed
Statistical analysis was carried out using SPSS 21.0. Data are a significant increase in all groups at the end of the experi-
expressed as the mean ± standard deviation (SD) and were ana- ment, but again, the HFD group showed the highest BMI
lyzed by one- or two-way ANOVA with post hoc analysis followed among the three groups, whereas it did not significantly differ
by Tukey’s or Duncan’s tests. GraphPad Prism 5 (GraphPad, La between NFD and HAOS (Fig. 1D). HFD significantly elevated
Jolla, CA, USA) was used to draw figures. Differences were con- the fasting blood glucose level (122.0 ± 15.1 mg dL−1 vs. 56.6 ±
sidered significant at p < 0.05. 13.8 mg dL−1 in NFD, p < 0.001), whereas AOS had a signifi-
cant suppressive effect (80.6 ± 17.8 mg dL−1, p < 0.05) (Fig. 1E).
Our data indicated that the zebrafish obesity model was suc-
3. Results and discussion cessfully established by five weeks of HFD. Our results clearly
indicated that dietary AOS has an anti-obesity effect as it atte-
3.1. Characterization of alginate oligosaccharide (AOS) nuated weight gain and BMI and decreased blood glucose
The chromatogram of sodium alginate showed two peaks, levels in HFD-fed zebrafish.
corresponding with average MWs of 351 727 Da, and 749 Da,
while the depolymerized sodium alginate (AOS) prepared 3.3. AOS suppresses lipogenesis and elevates energy
using bacterial alginate lyase showed three major peaks, expenditure in obese zebrafish
corresponding with average MWs of 167 471 Da, 796 Da, and To investigate the effects of HFD and the potentially regulatory
213 Da (Fig. S1A†), indicating that the enzyme had degraded effect of AOS on lipid metabolism, we evaluated the expression
the extended polymer structure of the alginate. The percentage of genes encoding leptin (LEPTIN), fatty acid synthase (FAS),
(%) of the area of each peak was 73.27%, 18.46%, and 8.27%, and sterol regulatory element binding proteins (SREBP1),
respectively, and it was found that the total alginate oligosac- which are lipid metabolism markers, by qRT-PCR. The results
charides usable were 26% in total fermented products (data are shown in Fig. 2A–C. As expected, HFD strongly induced the
not shown). Circular dichroic spectra of alginate showed a expression of LEPTIN (>80-fold), FAS (>20-fold increased), and
peak at 200.4 nm and a trough at 212.9 nm, while AOS showed SREBP1 (>2-fold), suggesting that HFD induced leptin pro-
a peak at 199.8 nm and a trough at 213.4 nm, respectively duction and insulin resistance, leading to gain weight and
(Fig. S1B†). Similarly, a previous study reported on poly-man- elevated blood glucose in zebrafish.39 LEPTIN was the most

This journal is © The Royal Society of Chemistry 2019 Food Funct.


View Article Online

Paper Food & Function


Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

Fig. 1 Changes in body weight, BMI, and fasting blood glucose in zebrafish on the different diets. (A–C) Changes in body weight during the five-
week feeding period and representative images of zebrafish at the start and the end of the experiment. (D) BMI and (E) fasting blood glucose in adult
zebrafish after 5 weeks of NFD, HFD, and HAOS.

strongly upregulated gene in HFD-fed zebrafish. In agreement plementation strongly enhanced UCP1 expression (by 18.4-fold
with our results, Simonds et al. reported that leptin plays an as compared to NFD, Fig. 2H). AMPK plays important roles in
important role in the increase in blood pressure and body energy homeostasis regulation and stimulates fatty acid
weight associated with obesity in mice.40 In humans, leptin β-oxidation. Thus, activated AMPK is involved in the inhibition
mRNA concentrations in adipose tissue and serum positively of lipogenesis or elevation of lipolysis, inducing weight loss.41
correlate with the amount of fat mass, and fasting or weight PGC-1α, as a pivotal regulator of mitochondrial biogenesis and
loss lowers leptin levels.39 In line with our findings, overfeed- oxidative metabolism, is another key regulator of cellular
ing strongly induced FAS and SREBP1 expression in adult zeb- energy metabolism.42 SIRT1 is considered a critical metabolic
rafish.22 AOS significantly suppressed LEPTIN, FAS, and sensor that directly regulates a number of targets involved in
SREBP1 mRNA expression induced by HFD, indicating that lipid metabolism,43 whereas UCP1 is a predominant thermo-
AOS exerts anti-adipogenic activity. genic marker in brown adipose tissue.44 AMPK can increase
Elevating energy expenditure is one of the best approaches PGC-1α and SIRT1 activity and UCP1 expression and activity.45
to fight obesity.39 We evaluated whether AOS enhances whole- Hence, AMPKα, PGC-1α, SIRT1, and UCP1 form a metabolism-
body energy expenditure in HFD, by measuring the expression energy sensing network that stimulates energy expenditure to
of energy metabolism-related genes encoding AMP-activated prevent metabolic dysfunction.45 AMPKα expression and
protein kinase α (AMPKα), peroxisome proliferator-activated activity were measured by western blotting, which revealed
receptor gamma coactivator 1 α (PGC-1α), sirtuin (SIRT1), strong activation of AMPKα by dietary AOS (Fig. S3†), in line
acetyl-CoA carboxylase A (ACCA), and uncoupling protein 1 with the qRT-PCR results. Our results suggest that dietary AOS
(UCP1) by qRT-PCR. The results are shown in Fig. 2D–H. HFD supplementation in zebrafish has a significant impact on
dramatically reduced the expression of all biomarkers tested. AMPKα, UCP1, SIRT1, and PGC-1α expression and AMPK
Interestingly, AOS supplementation restored marker expression activity, which may explain its effect in preventing body fat
to levels similar to those in the NFD group. Notably, AOS sup- accumulation under HFD. Previous studies have also shown

Food Funct. This journal is © The Royal Society of Chemistry 2019


View Article Online

Food & Function Paper


Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

Fig. 2 Effect of HFD and AOS supplemented on the mRNA expression of target genes. AOS modulates mRNA expression of the lipid metabolism-
related genes FAS (A), LEPTIN (B), and SREBP1 (C), the energy metabolism-related genes AMPKα (D), PGC-1α (E), SIRT1 (F), ACCA (G), UCP1 (H), and
ATP2A1 (I). Results are means ± SD of 3–4 experiments. *p < 0.05, **p < 0.01, and ***p < 0.001.

that HFD profoundly decreases AMPKα, PGC-1α, SIRT1, and in all treatment groups. The data are shown in Fig. 3A–C,
UCP1 expression levels in C57BL/6 mice.42,46 As AMPKα, SIRT1, mRNA levels of all tested pro-inflammatory genes were
PGC-1α, and UCP1 are considered vital nutrients and energy increased in the HFD group compared to the NFD group. AOS
sensors, it is highly likely that AOS promotes energy expenditure significantly inhibited the expression of these pro-inflamma-
in HFD-fed zebrafish by elevating their activities. Thus, AOS is tory cytokines. These findings suggest that HFD induces
promising for nutritional or pharmacological interventions in inflammation, which may further induce obesity in zebrafish.
the treatment of obesity and metabolic disorders. Accordingly, previous in vivo studies have reported that the
inflammatory genes IL-1β, IL-6, MCP-1, and TNF-α are
3.4. AOS reverses HFD-induced inflammatory and apoptotic increased in HFD-induced obesity.46,47 A study on zebrafish
gene expression reported that obesity was strongly associated with upregulation
Given the impacts of inflammation on adipokine production, of the inflammation-related genes TNF-α, IL-1β, MMP2, and
we evaluated expression levels of inflammatory genes encoding MMP9.48 The suppressive effect of AOS on inflammatory
interleukins (IL-1β, IL-6), and tumor necrosis factor α (TNF-α) cytokine production may be associated with decreases in

This journal is © The Royal Society of Chemistry 2019 Food Funct.


View Article Online

Paper Food & Function


Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

Fig. 3 Effect of HFD and AOS supplemented on the mRNA expression of target genes. AOS modulates mRNA expression of the inflammatory cyto-
kines related genes IL-1β (A), IL-6 (B), and TNF-α (C), and the apoptosis-related genes BAX (D), BCL-2 (E), BAX/BCL-2 ratio (F), NR3C1 (G), TP53 (H),
and VDAC1 (I). Results are means ± SD of 3–4 experiments. *p < 0.05, **p < 0.01, and ***p < 0.001.

energy metabolic and lipogenic gene expression, as mentioned transcript levels in zebrafish after the five-week feeding period,
above.46 indicating it induced apoptosis. AOS supplementation signifi-
To evaluate the potentially inhibitory effect of AOS on HFD- cantly reversed HFD-induced apoptosis-related gene
induced apoptosis in zebrafish, we measured the expression of expression. AOS significantly up- and downregulated BCL2 and
the apoptosis-related genes BAX and BCL-2 and genes encod- BAX expression, respectively, leading to a significant decrease
ing the proapoptotic tumor protein p53 (TP53), voltage-depen- in the BAX/BCL2 ratio. BCL-2 is an important antiapoptotic
dent anion-selective channel protein 1 (VDAC1), and nuclear gene, whereas BAX is the most critical proapoptotic gene, and
receptor subfamily 3, group C, member 1 (NR3C1) by the BAX/BCL-2 ratio strongly contributes to apoptosis.49 As
qRT-PCR. As shown in Fig. 3D–H, HFD highly induced shown in Fig. 3G–I, AOS significantly attenuated the expression
expression of BAX, TP53, and NR3C1 and downregulated BCL-2 of NR3C1, TP53, and VDAC1, to levels observed in the NFD

Food Funct. This journal is © The Royal Society of Chemistry 2019


View Article Online

Food & Function Paper

group. BAX, TP53, and BCL-2 are apoptosis biomarkers, and and in vivo. Tusi et al. reported that AOS protected PC12 cells
VDAC1, an outer mitochondrial membrane, is also a key player from apoptosis by inducing BCL-2 expression and inhibiting
in apoptosis and is involved in cytochrome c release and inter- BAX expression and suppressing H2O2-induced caspase-3
actions with antiapoptotic genes/proteins.50 NR3C1 is a corti- activation.49
sol receptor that is induced in the hypothalamus–pituitary–
adrenal axis by stress.51 Apoptosis induction by HFD may 3.5. Comparative proteomics analysis of the effects of HFD
occur via oxidative stress as reactive oxygen species (ROS) are and AOS
massively produced in obesity.28,50 Our findings suggest that To elucidate the mechanisms of dietary AOS in suppressing
dietary AOS protects against HFD-induced apoptosis in zebra- obesity and modulating immuno-metabolic responses to HFD,
fish by modulating apoptosis-related genes. This is consistent samples from the three treatment groups were subjected to
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

with previous findings that AOS decreased apoptosis in vitro proteomic analysis. In total, 519 proteins were identified in the

Fig. 4 Venn diagram and heatmaps of significantly differentially expressed proteins based on their relative expression ratios in zebrafish on HFD and
HAOS. (A) Common proteins (146) between HFD vs. NFD and HAOS vs. HFD. (B) Common proteins (14) between HFD vs. NFD (downregulated) and
HAOS vs. HFD (upregulated). (C) Common proteins (132) between HFD vs. NFD (upregulated) and HAOS vs. HFD (downregulated). Ratios are shown
as log2 fold change, with upregulated proteins having expression >0.0, red; downregulated proteins having expression <0.0, green.

This journal is © The Royal Society of Chemistry 2019 Food Funct.


View Article Online

Paper Food & Function

three groups (Table S3-1†). Of these, 308 and 177 proteins were 3-phosphate dehydrogenase 2 (GAPDHS), pyruvate kinase-
significantly ( p < 0.05) differently expressed in HFD vs. ND and muscle, a (PKMA), and phosphoglycerate mutase 2 (PGAM2),
HAOS vs. HFD, respectively (Fig. 4A). Heatmaps based on the were upregulated in the HFD group, whereas enolase (ENO1b)
expression of these proteins are shown in Fig. 4B and C. and L-lactate dehydrogenase B–B chain (LDHBB) were
Remarkably, 146 proteins were found in common between decreased under HFD. AOS significantly reversed the
HFD vs. NFD and HAOS vs. HFD (Fig. 4A and Table S3-2†); of expression of these proteins (Fig. S4C†).
these, most (132 proteins) were upregulated by HFD and down- HFD strongly upregulated the expression of various ER-
regulated by AOS (Fig. 4C). Only 14 proteins were downregu- related proteins, especially, HSPA8, HSP90b1, HSC70, and
lated in HFD vs. NFD, and AOS treatment promoted the upre- HSPA5 by 4.4-, 3.3-, 1.9-, and 1.4-fold, respectively, when com-
gulation of these proteins (Fig. 4B). Remarkably, STOML2 was pared to NFD (Fig. S4C†), whereas these proteins were downre-
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

the most differentially regulated among the proteins identi- gulated by AOS. HSP70 and HSP90 are molecular chaperones
fied; it was induced by >16.5-fold by HFD (Table S3-2†). AOS whose expression is increased by various types of stress.49 We
significantly downregulated STOML2 by 0.52-fold when com- confirmed the proteomics expression data for HSP70 and
pared to the level in the HFD group (Table S3-2†). IPA analysis HSPA8 by western blotting (Fig. S3†). The significant upregula-
revealed that, among 146 common significantly differentially tion of all HSPs upon HFD may indicate that zebrafish try to
expressed proteins, which were related to cancer progression, adapt to the stress induced by HFD.52 GSEA showed that pro-
tumorigenesis, cancer metastasis, inflammation, and apopto- teins related to metabolic pathways were the most strongly
sis, as well as hepatic diseases and pathogenic diseases enriched among the proteins detected. Many of these proteins
(Table S4†). Network relationships and biological functions of are involved in both metabolic pathways and immune systems.
the proteins identified were analyzed by STRING analysis. STOML2 has been shown to be involved in various metabolic
Among the 146 common proteins, 51 proteins strongly ( p < process (Table 1). Based on the proteomic data, STOML2 was
0.05) interacted together and were differentially regulated predicted to be a central regulator in HFD-induced obesity and
between HFD and AOS treatments and may thus be considered was selected for further investigation of its role in immuno-
as potential biomarkers of immune regulation and the anti- metabolic systems.
obesity mechanism of AOS in zebrafish (Fig. S4A†). In
addition, according to IPA and KEGG pathway analysis 3.6. STOML2 regulates crosstalk between immune systems
(Table S5 and Fig. S4B†), most of the identified proteins were and metabolic pathways to control HFD-induced obesity
assigned to two major groups: metabolic pathways (glycolysis/ First, we confirmed the relative mRNA expression of STOML2
gluconeogenesis) and the immune system (unfolded protein in zebrafish by qRT-PCR. HFD significantly increased STOML2
response, mitochondrial dysfunction, endoplasmic reticulum, expression (8.9-fold) as compared to NFD. Meanwhile, AOS
etc.). Remarkably, STOML2 seems to act as a central mediator strongly inhibited this increase (1.4-fold increase in HAOS vs.
connecting metabolic pathways and the immune system NFD) (Fig. 5). This was in accordance with the results obtained
(Fig. S4A†). Glycolysis/gluconeogenesis is a fundamental and by proteomics analysis. Similarly, STOML2 reportedly was
highly conserved metabolic pathway in most organisms. Five upregulated in skeletal muscle biopsies from obese as com-
major glycolytic proteins (common proteins of glucose metab- pared to non-obese subjects.53 While STOML2 expression is
olism, gluconeogenesis, glycolysis, and glycogenolysis), includ- induced under different conditions, the mechanism under-
ing aldolase b (ALDOB), aldolase c (ALDOCB), glyceraldehyde- lying inhibition/activation of STOML2 is still unknown.

Table 1 Most enriched gene/gene sets determined by GSEA (http://software.broadinstitute.org) (with only FDR q-values < 0.05) in zebrafish on
HFD with AOS supplementation

FDR
Gene/gene set p-Value q-value Molecules
−12 −8
GO small molecule 1.11 × 10 1.98 × 10 HSPA8, AHCY, ATP5A1, GAPDHS, PGAM2, ALDOB, ATP5B, COX6C,
metabolic process [1767] STOML2, BHMT, P4HB, ALDH4A1, APOA2, DDC, CS, GPD2, ME3
GO glycosyl compound 4.79 × 10−12 4.27 × 10−8 HSPA8, AHCY, ATP5A1, GAPDHS, PGAM2, ALDOB, ATP5B, COX6C,
metabolic process [368] STOML2, BHMT
GO purine containing compound 9.37 × 10−12 5.56 × 10−8 HSPA8, AHCY, ATP5A1, GAPDHS, PGAM2, ALDOB, ATP5B, COX6C,
metabolic process [394] STOML2, BHMT
GO organonitrogen compound 1.91 × 10−11 6.81 × 10−8 HSPA8, AHCY, ATP5A1, GAPDHS, PGAM2, ALDOB, ATP5B, COX6C,
metabolic process [1796] STOML2, BHMT, P4HB, ALDH4A1, APOA2, DDC, RPL7, GSTP1
GO nucleoside triphosphate 1.03 × 10−10 2.3 × 10−7 HSPA8, ATP5A1, GAPDHS, PGAM2, ALDOB, ATP5B, COX6C, STOML2
metabolic process [228]
GO nucleoside monophosphate 1.5 × 10−10 2.45 × 10−7 HSPA8, ATP5A1, GAPDHS, PGAM2, ALDOB, ATP5B, COX6C, STOML2
metabolic process [239]

The p-value and FDR q-value were calculated using Fisher’s exact test by GSEA. FDR: false discovery rate. Only the most-significant terms are
shown.

Food Funct. This journal is © The Royal Society of Chemistry 2019


View Article Online

Food & Function Paper


Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

Fig. 5 Effects of inhibitor/activator treatments on the expression of biomarkers in zebrafish. (A) Effect of AICAR treatment on mRNA expression of
AMPKα, PGC-1α, and STOML2 and FAS, LEPTIN, and IL-1β. (B) Effect of U0126 treatment on mRNA expression of AKT1, ERK1, and STOML2, and FAS,
LEPTIN, and AMPKα. (C) Effect of KNK437 treatment on the expression of HSPA5, HSP90b1, and STOML2, and FAS, LEPTIN, and AMPKα. (D) Effect of
LPS treatment on mRNA expression of IFN-γ, IL-6, and STOML2, and FAS, LEPTIN, and AMPKα. Results are means ± SD of 3–4 experiments. NS, not
significant *p < 0.05, **p < 0.01, and ***p < 0.001.

If STOML2 is a central factor that plays important roles in inhibitors/activators of these pathways (AMPKα/SIRT1/PGC1α,
preventing obesity by targeting multiple pathways in immuno- energy expenditure signaling; HSP signaling; AKT/MAPK/ERK
metabolic interaction, STOML2 expression should be regulated signaling). Remarkably, U0126, a specific MEK/ERK inhibi-
by these specific inhibitors or activators. To confirm whether a tor,54 the most strongly reduced the HFD-induced STOML2
possible crosstalk among those pathways modulating lipogen- response (∼20-fold decrease as compared to HFD without
esis is linked to the regulation of STOML2, we used various inhibitor treatment, Fig. 5C). Similarly, AICAR, an AMPK

This journal is © The Royal Society of Chemistry 2019 Food Funct.


View Article Online

Paper Food & Function


Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

Fig. 6 Proposed effects of dietary AOS on high fat diet-induced obesity and pathophysiological disorders in zebrafish. AOS regulates immuno-
metabolic systems by regulating pro-inflammatory cytokines, inhibiting apoptosis, and modulating energy expenditure and lipid metabolism by regu-
lating STOML2 in high fat diet-induced obesity in zebrafish.

activator41 suppressed STOML2 expression by about 10-fold as brane protein in human red blood cells that has been recently
compared to HFD without AICAR treatment (Fig. 5B). KNK437, revealed to be associated with a variety of diseases.57 Increased
a novel inhibitor of HSP,55 and LPS-induced inflammation12,56 STOML2 expression has been associated with increased risk
in zebrafish had a positive effect on STOML2 expression. As for asthma, suggesting that STOML2 may have a pro-inflamma-
predicted, KNK437 treatment in the HFD group inhibited tory effect in asthma.58 Song et al. have reported that downre-
STOML2 expression (∼5.5-fold decrease, Fig. 5C), whereas LPS gulation of STOML2 reduces NF-κB transactivity and inhibits
treatment for 3 days in the NFD group highly induced the the NF-κB pathway by downregulating of BCL-xL, TNF-α, MYC,
expression of inflammatory cytokines (IL-6 and IFN-γ) and CCND1, IL-6, and IL-8.57 Recently, Yang et al. found that
STOML2 (>3-fold vs. NFD, Fig. 5D). To evaluate whether STOML2 directly affected annexin A2 and β-catenin
STOML2 may act as a regulator of lipogenesis and thermogen- expression.27 Based on our findings in the current study, we
esis, we measured the expression AMPKα, PGC-1α, UCP1, ACCA, suggest that the changes in the immuno-metabolic system in
FAS, and LEPTIN, biomarkers of metabolic abnormalities, after zebrafish in response to HFD-induced obesity are regulated by
the above treatments. The transcriptional levels of all these STOML2 (Fig. 6). Recent evidence suggests mitochondria
markers were strongly correlated with STOML2 expression provide a powerful strategy to modulate energy homeostasis as
(Fig. 5B–D and Fig. S5†). In line with decreased STOML2 tran- well as metabolic health for the treatment of obesity and meta-
scription, FAS and LEPTIN expression was reduced upon treat- bolic disorders.28,59 Even though the exact function and
ment with AICAR, KNK437, and U0126 (Fig. 5A–C). In contrast, mechanism of STOML2 in metabolic syndromes remain
LPS induced the upregulation of STOML2, as well as of FAS unclear, our study provided evidence that STOML2 might be a
and LEPTIN (Fig. 5D). The energy expenditure-related genes promising novel target in the treatment of immuno-metabolic
AMPKα, PGC-1α, and UCP1 showed similar expression patterns disorders, as such obesity (Fig. 6).
under all treatments in correlation to STOML2 expression
(Fig. 5A–D and Fig. S5†). Remarkably, AOS mimicked the
actions of several molecules, such as AICAR, KNK437, and 4. Conclusions
U0126, which strongly activated energy expenditure and inhib-
ited STOML2 expression in zebrafish, and this may represent a This study is the first to report that AOS significantly prevents
novel mechanism of action for anti-obesity drugs (Fig. 6). HFD-induced immuno-metabolic dysfunction in zebrafish.
STOML2, a member of the human stomatin family, is a mem- Dietary AOS modulated a range of zebrafish genes and proteins

Food Funct. This journal is © The Royal Society of Chemistry 2019


View Article Online

Food & Function Paper

related to immuno-metabolic systems by inhibiting STOML2, oxidant capacity and intestinal digestion-absorption func-
thus maintaining/improving health in zebrafish fed an HFD. tion in weaned pigs, Anim. Feed Sci. Technol., 2017, 234,
Our findings pave the way for future dietary interventions and 118–127.
clinical recommendations. Especially, STOML2, identified in 6 M. Szekalska, A. Puciłowska, E. Szymańska, P. Ciosek and
this study as a key regulator, warrants further study to develop K. Winnicka, Alginate: Current use and future perspectives
a potential treatment for metabolic disorders. in pharmaceutical and biomedical applications,
Int. J. Polym. Sci., 2016, 2016, 1–17.
7 M. Falkeborg, L. Z. Cheong, C. Gianfico, K. M. Sztukiel,
Abbreviations K. Kristensen, M. Glasius, X. Xu and Z. Guo, Alginate oligo-
saccharides: Enzymatic preparation and antioxidant prop-
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

AOS Alginate oligosaccharide erty evaluation, Food Chem., 2014, 164, 185–194.
HFD High-fat-diet 8 J. Wan, J. Zhang, D. Chen, B. Yu, Z. Huang, X. Mao,
NFD Normal diet P. Zheng, J. Yu and J. He, Alginate oligosaccharide
enhances intestinal integrity of weaned pigs through alter-
ing intestinal inflammatory responses and antioxidant
Author contributions status, RSC Adv., 2018, 8, 13482–13492.
9 A. Tøndervik, H. Sletta, G. Klinkenberg, C. Emanuel,
D. K. designed the study; T. V. C. performed most of the experi- L. C. Powell, M. F. Pritchard, S. Khan, K. M. Craine,
ments and statistical analysis; T. V. C., and D. K. wrote the E. Onsøyen, P. D. Rye, C. Wright, D. W. Thomas and
manuscript; S. Y. C., and J. K., conducted proteomic analysis; K. E. Hill, Alginate oligosaccharides inhibit fungal cell
T. V. C., and S. Y. C., contributed to data organization, or ana- growth and potentiate the activity of antifungals against
lysis; and J. K., and D. K. supervised the study. All authors con- Candida and Aspergillus Spp., PLoS One, 2014, 9, e112518.
tributed to comments and revision on the manuscript and 10 J. Wan, J. Zhang, D. Chen, B. Yu, X. Mao, P. Zheng, J. Yu,
approved the final manuscript. J. Luo and J. He, Alginate oligosaccharide-induced intesti-
nal morphology, barrier function and epithelium apoptosis
modifications have beneficial effects on the growth per-
Conflicts of interest formance of weaned pigs, J. Anim. Sci. Biotechnol., 2018, 9,
58–69.
The authors declared no conflicts of interest.
11 J.-J. Guo, L.-L. Ma, H.-T. Shi, J.-B. Zhu, J. Wu, Z.-W. Ding,
Y. An, Y.-Z. Zou and J.-B. Ge, Alginate oligosaccharide pre-
vents acute doxorubicin cardiotoxicity by suppressing oxi-
Acknowledgements dative stress and endoplasmic reticulum-mediated apopto-
This study was supported by a grant from the Korea Basic sis, Mar. Drugs, 2016, 14, 231–243.
Science Institute (No. C39712). 12 R. Zhou, X. Shi, Y. Gao, N. Cai, Z. Jiang and X. Xu, Anti-
inflammatory activity of guluronate oligosaccharides
obtained by oxidative degradation from alginate in lipopo-
References lysaccharide-activated murine macrophage RAW 264.7
Cells, J. Agric. Food Chem., 2015, 63, 160–168.
1 L. K. Ma, B. Zhang, S. G. Deng and C. Xie, Comparison of 13 X. Hu, X. Jiang, H. Hwang, S. Liu and H. Guan, Antitumour
the cryoprotective effects of trehalose, alginate, and its activities of alginate-derived oligosaccharides and their
oligosaccharides on peeled shrimp (Litopenaeus vannamei) sulphated substitution derivatives, Eur. J. Phycol., 2004, 39,
during frozen storage, J. Food Sci., 2015, 80, C540–C546. 67–71.
2 B. Zhang, H. Wu, H. Yang, X. Xiang, H. Li and S. Deng, 14 J. Chen, Y. Hu, L. Zhang, Y. Wang, S. Wang, Y. Zhang,
Cryoprotective roles of trehalose and alginate oligosacchar- H. Guo, D. Ji and Y. Wang, Alginate oligosaccharide DP5
ides during frozen storage of peeled shrimp (Litopenaeus exhibits antitumor effects in osteosarcoma patients follow-
vannamei), Food Chem., 2017, 228, 257–264. ing surgery, Front. Pharmacol., 2017, 8, 1–11.
3 H. Liu, Y. H. Zhang, H. Yin, W. X. Wang, X. M. Zhao and 15 S. Nakazono, K. Cho, S. Isaka, R. Abu, T. Yokose,
Y. G. Du, Alginate oligosaccharides enhanced Triticum aesti- M. Murata, M. Ueno, K. Tachibana, K. Hirasaka, D. Kim
vum L. tolerance to drought stress, Plant Physiol. Biochem., and T. Oda, Anti-obesity effects of enzymatically-digested
2013, 62, 33–40. alginate oligomer in mice model fed a high-fat-diet, Bioact.
4 Y. Zhang, H. Yin, X. Zhao, W. Wang, Y. Du, A. He and Carbohydr. Diet. Fibre, 2016, 7, 1–8.
K. Sun, The promoting effects of alginate oligosaccharides 16 K. a. Gabor, M. F. Goody, W. K. Mowel, M. E. Breitbach,
on root development in Oryza sativa L. mediated by auxin R. L. Gratacap, P. E. Witten and C. H. Kim, Influenza A
signaling, Carbohydr. Polym., 2014, 113, 446–454. virus infection in zebrafish recapitulates mammalian infec-
5 J. Wan, J. Zhang, D. Chen, B. Yu and J. He, Effects of algi- tion and sensitivity to anti-influenza drug treatment, Dis.
nate oligosaccharide on the growth performance, anti- Models Mech., 2014, 7, 1227–1237.

This journal is © The Royal Society of Chemistry 2019 Food Funct.


View Article Online

Paper Food & Function

17 B. Novoa, A. Romero, V. Mulero, I. Rodríguez, I. Fernández negative predictor of survival, Clin. Proteomics, 2016, 13, 1–
and A. Figueras, Zebrafish (Danio Rerio) as a model for the 12.
study of vaccination against viral haemorrhagic septicemia 30 S. K. Garg, H. Maurer, K. Reed and R. Selagamsetty,
virus (VHSV), Vaccine, 2006, 24, 5806–5816. Diabetes and cancer: Two diseases with obesity as a
18 C. Medina and J. L. Royo, Zebrafish as a model organism common risk factor, Diabetes, Obes. Metab., 2014, 16, 97–
to study host-pathogen interactions, Methods, 2013, 62, 110.
241–245. 31 X. Lu and H. Zhu, Tube-Gel digestion, Mol. Cell. Proteomics,
19 N. Hachicho, S. Reithel, A. Miltner, H. J. Heipieper, 2005, 4, 1948–1958.
E. Küster and T. Luckenbach, Body Mass Parameters, Lipid 32 B. Vaidya, S. Y. Cho, K. S. Oh, S. H. Kim, Y. O. Kim,
profiles and protein contents of zebrafish embryos and E. H. Jeong, T. T. Nguyen, S. H. Kim, I. S. Kim, J. Kwon and
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

effects of 2,4-dinitrophenol exposure, PLoS One, 2015, 10, D. Kim, Effectiveness of periodic treatment of quercetin
1–19. against influenza A virus H1N1 through modulation of
20 T. Oka, Y. Nishimura, L. Zang, M. Hirano, Y. Shimada, protein expression, J. Agric. Food Chem., 2016, 64, 4416–
Z. Wang, N. Umemoto, J. Kuroyanagi, N. Nishimura and 4425.
T. Tanaka, Diet-induced obesity in zebrafish shares 33 J. G. Burniston, J. Connolly, H. Kainulainen, S. L. Britton
common pathophysiological pathways with mammalian and L. G. Koch, Profiling of skeletal muscle using high-
obesity, BMC Physiol., 2010, 10, 21–33. definition mass spectrometry, Proteomics, 2014, 14, 2339–
21 S. Meguro, T. Hasumura and T. Hase, Body fat accumu- 2344.
lation in zebrafish is induced by a diet rich in fat and 34 A. I. Saeed, V. Sharov, J. White, J. Li, W. Liang,
reduced by supplementation with green tea extract, PLoS N. Bhagabati, J. Braisted, M. Klapa, T. Currier,
One, 2015, 10, e0120142. M. Thiagarajan, A. Sturn, M. Snuffin, A. Rezantsev,
22 K. Landgraf, S. Schuster, A. Meusel, A. Garten, T. Riemer, D. Popov, A. Ryltsov, E. Kostukovich, I. Borisovsky, Z. Liu,
D. Schleinitz, W. Kiess and A. Körner, Short-term overfeed- A. Vinsavich, V. Trush and J. Quackenbush, TM4: A free,
ing of zebrafish with normal or high-fat diet as a model for open-source system for microarray data management and
the development of metabolically healthy versus unhealthy analysis, BioTechniques, 2003, 34, 374–378.
obesity, BMC Physiol., 2017, 17, 4–13. 35 A. Subramanian, P. Tamayo, V. K. Mootha, S. Mukherjee,
23 W. Cao, B. Zhang, J. Li, Y. Liu, Z. Liu and B. Sun, SLP-2 B. L. Ebert, M. A. Gillette, A. Paulovich, S. L. Pomeroy,
overexpression could serve as a prognostic factor in node T. R. Golub, E. S. Lander and J. P. Mesirov, Gene set enrich-
positive and HER2 negative breast cancer, Pathology, 2011, ment analysis: A knowledge-based approach for interpret-
43, 713–718. ing genome-wide expression profiles, Proc. Natl. Acad.
24 L. Zhang, F. Ding, W. Cao, Z. Liu, W. Liu, Z. Yu, Y. Wu, Sci. U. S. A., 2005, 102, 15545–15550.
W. Li, Y. Li and Z. Liu, Stomatin-like protein 2 is over- 36 M. Chhatbar, R. Meena, K. Prasad and A. K. Siddhanta,
expressed in cancer and involved in regulating cell growth Microwave assisted rapid method for hydrolysis of sodium
and cell adhesion in human esophageal squamous cell car- alginate for M/G ratio determination, Carbohydr. Polym.,
cinoma, Clin. Cancer Res., 2006, 12, 1639–1646. 2009, 76, 650–656.
25 G. Hu, J. Zhang, F. Xu, H. Deng, W. Zhang, S. Kang and 37 T. Muramatsu, Change in Circular dichroic spectra of the
W. Liang, Stomatin-like protein 2 inhibits cisplatin- substrate caused by alginate lyase, Agric. Biol. Chem., 1986,
induced apoptosis through MEK/ERK signaling and the 50, 1311–1313.
mitochondrial apoptosis pathway in cervical cancer cells, 38 T. Tainaka, Y. Shimada, J. Kuroyanagi, L. Zang, T. Oka,
Cancer Sci., 2018, 109, 1357–1368. Y. Nishimura, N. Nishimura and T. Tanaka, Transcriptome
26 H. Deng, Y. Deng, F. Liu, J. Chen, Z. Li, K. Zhao, X. Guan analysis of anti-fatty liver action by campari tomato using a
and W. Liang, Stomatin-like protein 2 is overexpressed in zebrafish diet-induced obesity model, Nutr. Metab., 2011, 8,
cervical cancer and involved in tumor cell apoptosis, Oncol. 88–98.
Lett., 2017, 14, 6355–6364. 39 A. A. Van Der Klaauw and I. S. Farooqi, The Hunger Genes:
27 C.-T. Yang, J.-M. Li, L.-F. Li, Y.-S. Ko and J.-T. Chen, Pathways to obesity, Cell, 2015, 161, 119–132.
Stomatin-like protein 2 regulates survivin expression in 40 S. E. Simonds, J. T. Pryor, E. Ravussin, F. L. Greenway,
non-small cell lung cancer cells through β-catenin signal- R. Dileone, A. M. Allen, J. Bassi, J. K. Elmquist,
ing pathway, Cell Death Dis., 2018, 9, 425–435. J. M. Keogh, E. Henning, M. G. Myers, J. Licinio,
28 N. K. Bhatraju and A. Agrawal, Mitochondrial dysfunction R. D. Brown, P. J. Enriori, S. O’Rahilly, S. M. Sternson,
linking obesity and asthma, Ann. Am. Thorac. Soc., 2017, 14, K. L. Grove, D. C. Spanswick, I. S. Farooqi and
S368–S373. M. A. Cowley, Leptin mediates the increase in blood
29 J. F. Fahrmann, D. Grapov, B. S. Phinney, C. Stroble, pressure associated with obesity, Cell, 2014, 159, 1404–
B. C. DeFelice, W. Rom, D. R. Gandara, Y. Zhang, O. Fiehn, 1416.
H. Pass and S. Miyamoto, Proteomic profiling of lung 41 A. Zhang, H. Sun and X. Wang, Emerging role and recent
adenocarcinoma indicates heightened DNA repair, anti- applications of metabolomics biomarkers in obesity
oxidant mechanisms and identifies LASP1 as a potential disease research, RSC Adv., 2017, 7, 14966–14973.

Food Funct. This journal is © The Royal Society of Chemistry 2019


View Article Online

Food & Function Paper

42 X. Zhang, Q.-X. Zhang, X. Wang, L. Zhang, W. Qu, B. Bao, 51 G. Gioacchini, E. Giorgini, I. Olivotto, F. Maradonna,
C.-A. Liu and J. Liu, Dietary luteolin activates browning D. L. Merrifield and O. Carnevali, The influence of probio-
and thermogenesis in mice through an AMPK/PGC1α tics on zebrafish Danio rerio innate immunity and hepatic
pathway-mediated mechanism, Int. J. Obes., 2016, 40, stress, Zebrafish, 2014, 11, 98–106.
1841–1849. 52 P. Dowling, P. Meleady, A. Dowd, M. Henry, S. Glynn and
43 T. T. Schug and X. Li, Sirtuin 1 in lipid metabolism and M. Clynes, Proteomic analysis of isolated membrane frac-
obesity, Ann. Med., 2011, 43, 198–211. tions from superinvasive cancer cells, Biochim. Biophys.
44 B. Shan, X. Wang, Y. Wu, C. Xu, Z. Xia, J. Dai, M. Shao, Acta, Proteins Proteomics, 2007, 1774, 93–101.
F. Zhao, S. He, L. Yang, M. Zhang, F. Nan, J. Li, J. Liu, 53 L. Formentini, A. J. Ryan, M. Gálvez-Santisteban, L. Carter,
J. Liu, W. Jia, Y. Qiu, B. Song, J.-D. J. Han, L. Rui, P. Taub, J. D. Lapek, D. J. Gonzalez, F. Villarreal,
Published on 04 July 2019. Downloaded by KEAN UNIVERSITY on 7/23/2019 8:33:54 AM.

S.-Z. Duan and Y. Liu, The metabolic ER stress sensor T. P. Ciaraldi, J. M. Cuezva and R. R. Henry, Mitochondrial
IRE1α suppresses alternative activation of macrophages H+-ATP synthase in human skeletal muscle: Contribution
and impairs energy expenditure in obesity, Nat. Immunol., to dyslipidaemia and insulin resistance, Diabetologia, 2017,
2017, 18, 519–529. 60, 2052–2065.
45 C. Cantó and J. Auwerx, PGC-1α, SIRT1 and AMPK, an 54 S. L. Hwang, Y. T. Jeong, X. Li, Y. D. Kim, Y. Lu,
Energy sensing network that controls energy expenditure, Y. C. Chang, I. K. Lee and H. W. Chang, Inhibitory cross-
Curr. Opin. Lipidol., 2009, 20, 98–105. talk between the AMPK and ERK pathways mediates endo-
46 J. Dong, X. Zhang, L. Zhang, H.-X. Bian, N. Xu, B. Bao and plasmic reticulum stress-induced insulin resistance in skel-
J. Liu, Quercetin reduces obesity-associated ATM infiltra- etal muscle, Br. J. Pharmacol., 2013, 169, 69–81.
tion and inflammation in mice: A mechanism including 55 E. H. Jeong, B. Vaidya, S. Y. Cho, M. A. Park,
AMPKα1/SIRT1, J. Lipid Res., 2014, 55, 363–374. K. Kaewintajuk, S. R. Kim, M. J. Oh, J. S. Choi, J. Kwon and
47 A. Lama, C. Pirozzi, M. P. Mollica, G. Trinchese, F. Di D. Kim, Identification of regulators of the early stage of
Guida, G. Cavaliere, A. Calignano, G. Mattace Raso, viral hemorrhagic septicemia virus infection during curcu-
R. Berni Canani and R. Meli, Polyphenol-rich virgin olive min treatment, Fish Shellfish Immunol., 2015, 45, 184–193.
oil reduces insulin resistance and liver inflammation and 56 G. Forn-Cuní, M. Varela, C. M. Fernández-Rodríguez,
improves mitochondrial dysfunction in high-fat diet fed A. Figueras and B. Novoa, Liver immune responses to
rats, Mol. Nutr. Food Res., 2017, 61, 1600418. inflammatory stimuli in a dietinduced obesity model of
48 C.-Y. Chu, C.-F. Chen, R. S. Rajendran, C.-N. Shen, zebrafish, J. Endocrinol., 2015, 224, 159–170.
T.-H. Chen, C.-C. Yen, C.-K. Chuang, D.-S. Lin and 57 L. Song, L. Liu, Z. Wu, C. Lin, T. Dai, C. Yu, X. Wang,
C.-D. Hsiao, Overexpression of Akt1 enhances adipogenesis J. Wu, M. Li and J. Li, Knockdown of stomatin-like protein
and leads to lipoma formation in zebrafish, PLoS One, 2 (STOML2) reduces the invasive ability of glioma cells
2012, 7, e36474. through inhibition of the NF-KB/MMP-9 pathway, J. Pathol.,
49 S. K. Tusi, L. Khalaj, G. Ashabi, M. Kiaei and 2012, 226, 534–543.
F. Khodagholi, Alginate oligosaccharide protects against 58 J. A. Revez, M. C. Matheson, J. Hui, S. Baltic, A. James,
endoplasmic reticulum- and mitochondrial-mediated apop- J. W. Upham, S. Dharmage, P. J. Thompson, N. G. Martin,
totic cell death and oxidative stress, Biomaterials, 2011, 32, J. L. Hopper and M. A. R. Ferreira, Identification of
5438–5458. STOML2 as a putative novel asthma risk gene associated
50 S. Weisthal, N. Keinan, D. Ben-Hail, T. Arif and V. Shoshan- with IL6R, Allergy, 2016, 71, 1020–1030.
Barmatz, Ca2+-mediated regulation of VDAC1 expression 59 M. Ost, S. Keipert and S. Klaus, Targeted mitochondrial
levels is associated with cell death induction, Biochim. uncoupling beyond UCP1 – the fine line between death and
Biophys. Acta, Mol. Cell Res., 2014, 1843, 2270–2281. metabolic health, Biochimie, 2017, 134, 77–85.

This journal is © The Royal Society of Chemistry 2019 Food Funct.

You might also like