Journal Pre-proof Health benefits of fucoxanthin in the prevention of chronic diseases
Minkyung Bae, Mi-Bo Kim, Young-Ki Park, Ji-Young Lee PII:
S1388-1981(20)30010-X
DOI:
https://doi.org/10.1016/j.bbalip.2020.158618
Reference:
BBAMCB 158618
To appear in:
BBA - Molecular and Cell Biology of Lipids
Received date:
6 November 2019
Revised date:
5 January 2020
Accepted date:
7 January 2020
Please cite this article as: M. Bae, M.-B. Kim, Y.-K. Park, et al., Health benefits of fucoxanthin in the prevention of chronic diseases, BBA - Molecular and Cell Biology of Lipids(2020), https://doi.org/10.1016/j.bbalip.2020.158618
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
© 2020 Published by Elsevier.
Journal Pre-proof Health benefits of fucoxanthin in the prevention of chronic diseases
Minkyung Bae1, Mi-Bo Kim1, Young-Ki Park1, Ji-Young Lee1,2 1
Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut 06269,
USA Corresponding author: Ji-Young Lee, Ph.D. Fax: (860) 486-3674
Email:
[email protected]
of
Phone: (860) 486-2242
ro
2
-p
Running title: The health benefits of fucoxanthin
Abbreviations: AMPK, AMP-activated protein kinase; CPT-1, carnitine palmitoyl transferase 1;
re
C/EBPα, CCAAT/enhancer-binding protein alpha; CDK, cyclin-dependent kinase; COX-2,
lP
cyclooxygenase-2; FAS, fatty acid synthase; Fe-NTA, ferric nitrilotriacetate; G6PD, glucose-6-
na
phosphate dehydrogenase; GLUT4, glucose-transporter 4; GPDH, glycerol-3-phosphate dehydrogenase; HSCs, hepatic stellate cells; iNOS, inducible nitric oxide synthase; IL-1β,
ur
interleukin-1β; LPS, lipopolysaccharide; MCP-1, monocyte chemoattractant protein-1; ME,
Jo
malic enzyme; MMP, matrix metalloproteinase; NO, nitric oxide; NAFLD, nonalcoholic fatty liver disease; NF-κB, nuclear factor-κB; PI3K, phosphatidylinositol-3-kinase; PPARα, peroxisome proliferator-activated receptor α; PGE2, prostaglandin E2; ROS, reactive oxygen species; SR-BI, scavenger receptor class B, type 1; SREBP-1c, sterol regulatory element-binding protein 1c; TGFβ1, transforming growth factor β1; TNF-α, tumor necrosis factor-α; UCP-1, uncoupling protein 1
1
Journal Pre-proof Abstract Fucoxanthin is a xanthophyll carotenoid abundant in macroalgae, such as brown seaweeds. When fucoxanthin is consumed, it can be esterified or hydrolyzed to fucoxanthinol in the gastrointestinal tract and further converted into amarouciaxanthin A in the liver. It has a unique chemical structure that confers its biological effects. Fucoxanthin has a strong antioxidant capacity by scavenging singlet molecular oxygen and free radicals. Also, it exerts an anti-
of
inflammatory effect. Studies have demonstrated potential health benefits of fucoxanthin for the
ro
prevention of chronic diseases, such as cancer, obesity, diabetes mellitus, and liver disease.
-p
Animal studies have shown that fucoxanthin supplementation has no adverse effects. However,
re
investigation of the safety of fucoxanthin consumption in humans is lacking. Clinical trials are required to assess the safety of fucoxanthin in conjunction with the study of mechanisms by
lP
which fucoxanthin exhibits its health benefits. This review focuses on current knowledge of
na
metabolism and functions of fucoxanthin with its potential health benefits.
Jo
ur
Keywords: Fucoxanthin; Carotenoid; Brown seaweed; Antioxidant; Anti-inflammation
2
Journal Pre-proof Introduction Fucoxanthin is a xanthophyll carotenoid abundant in macroalgae, such as brown seaweeds, and microalgae [1]. Several edible brown algae, including Sargassam fusiforme, Laminaria japonica, Undaria pinnatifida, and Padina tetrastromatica, are consumed in SouthEast Asia and European countries [2]. The brown algae are good sources of fucoxanthin [3], however, diatoms, such as Phaeodactylum tricornutum, are preferred sources of fucoxanthin in
of
the food industry because of their higher fucoxanthin content and extraction efficiency with
ro
shorter growth cycle than those of macroalgae [4].
-p
Fucoxanthin has a unique chemical structure that contains an allenic bond, epoxy,
re
hydroxyl, carbonyl, and carboxyl groups in the molecule (Figure 1). Among ~700 carotenoids found in nature, only about 40 carotenoids have an allenic bond [3], which may confer a free
lP
radical scavenging activity [5]. It is known that fucoxanthin has a potent antioxidant capacity by
na
scavenging singlet molecular oxygen and free radicals [6]. However, fucoxanthin is unstable, and it can be easily degraded by heating, aerial exposure, or illumination [7].
ur
The primary metabolites of fucoxanthin are fucoxanthinol and amarouciaxanthin A.
Jo
Fucoxanthin can be hydrolyzed to fucoxanthinol in the gastrointestinal tract and further converted into amarouciaxanthin A in the liver (Figure 2) [8, 9]. [8]In animal studies, no toxicity of fucoxanthin was observed [12, 13]. Although clinical trials of fucoxanthin supplementation to assess its safety are lacking, the Food and Drug Administration allowed fucoxanthin extracted from alga, Phaeodactylum tricornutum, as a new dietary ingredient that can be consumed at a level of 3 mg daily with no time limit or 5 mg fucoxanthin daily for up to 90 days [14]. Studies have demonstrated that fucoxanthin may be used to prevent or treat chronic diseases, such as obesity, diabetes mellitus, and cancer. This review summarizes current
3
Journal Pre-proof knowledge of functions and potential health benefits of fucoxanthin, including anti-inflammatory, anti-cancer, anti-obesity, anti-diabetic, and hepatoprotective effects.
Chemical structure and antioxidant capacity Fucoxanthin is one of the carotenoids that contain an allenic bond in their molecules [11]. The major allenic carotenoids include fucoxanthin in brown algae or diatoms, peridinin in
of
dinoflagellates, and neoxanthin in plants and algae [15]. In addition to an allenic bond,
ro
fucoxanthin has epoxy, hydroxyl, carbonyl, and carboxyl groups [16].
-p
Carotenoids, including fucoxanthin, are known to have an antioxidant capacity that can
re
scavenge singlet molecular oxygen (1O2) and peroxyl radicals [17]. Singlet oxygen quenching by carotenoids mostly depends on physical quenching, i.e., transferring energy between two
lP
molecules [17, 18]. The energy of singlet oxygen is transferred to conjugated double bonds in the
na
central chain of carotenoids [11], leading to a triplet unreactive ground state of oxygen and a triplet excited state of the carotenoid molecules. The excited carotenoids return to a ground state
ur
by dissipating energy into the environment. During physical quenching, there are no chemical
Jo
reactions between singlet molecular oxygen and a carotenoid. Thus, carotenoids are not oxidized or consumed by the reaction, and therefore they can be reused. The efficiency of singlet oxygen quenching by carotenoids increases with the number of conjugated double bonds [19]. Carotenoids also scavenge free radicals. The ability of carotenoids to scavenge free radicals depends on the presence of functional groups with increasing polarity, such as carbonyl and hydroxyl groups, in their terminal rings [20]. Studies have demonstrated that fucoxanthin decreases intracellular reactive oxygen species (ROS) levels, lipid peroxidation, and protein carbonyl contents that are induced by ferric nitrilotriacetate in murine embryonic hepatic BNL
4
Journal Pre-proof CL.2 cells [21]. Also, fucoxanthin decreases malondialdehyde levels in tributyltin-treated HepG2 cells, thus increasing cell viability [22]. In addition, fucoxanthin is known to improve the cell’s endogenous antioxidant defense system. Fucoxanthin increased mRNA and protein levels of glutamate-cysteine ligase catalytic subunit and glutathione synthetase, which are involved in glutathione synthesis, in human keratinocyte cell line HaCaT cells [23]. Furthermore, fucoxanthin markedly increased the
of
nuclear protein level of nuclear factor-erythroid 2 related factor 2, a transcription factor that
ro
regulates the expression of several antioxidant proteins, such as heme oxygenase-1 and
re
-p
NAD(P)H: quinine oxidoreductase 1, in mouse hepatic BNL CL.2 cells [24].
Metabolism of fucoxanthin
lP
Digestion, absorption, and transport of fucoxanthin
na
Carotenoids are absorbed in the small intestine, similar to dietary lipids and lipid-soluble vitamins [25]. It is known that carotenoid absorption occurs in the proximal half of the small
ur
intestine [26]. For the intestinal absorption of carotenoids, there are several essential steps.
Jo
Carotenoids should be released from the food matrix to be solubilized into mixed lipid micelles in the lumen for the entry into intestinal mucosal cells [27]. It has been suggested that carotenoids are absorbed into the enterocyte via facilitated diffusion through scavenger receptor class B, type 1 (SR-BI) [28]. Although not proven, SR-BI is a plausible carrier of fucoxanthin for its intestinal absorption. It has been demonstrated that fucoxanthin is esterified in human intestinal Caco-2 cells [10]. Therefore, esterified fucoxanthin is likely incorporated into chylomicrons with lipids similarly to other carotenoids for the systemic transport.
5
Journal Pre-proof Fucoxanthin metabolites When fucoxanthin is consumed, it can be esterified [10] or hydrolyzed to fucoxanthinol in the gastrointestinal tract by cholesterol esterase and lipase [8]. Fucoxanthinol can be further metabolized into amarouciaxanthin A through dehydrogenation and isomerization in the liver [9]. In a pharmacokinetic study, when a single dose of fucoxanthin (65 mg/kg body weight) was administered to rats, fucoxanthin was detected in the circulation at 30 min post-administration
of
and it reached its maximal concentration of 29 µg/L at ~8 h, while the level of fucoxanthinol in
ro
plasma was its maximum of 263 µg/L at 11 h after fucoxanthin administration [30]. The data
-p
indicate that large amount of fucoxanthin is converted into fucoxanthinol in the intestine.
re
In humans, after a single oral administration of 31 mg fucoxanthin [31] or daily consumption of 6.1 mg fucoxanthin for a week [32], fucoxanthinol, but not amarouciaxanthin A,
lP
was detected in the circulation. In mice, however, studies have shown that both fucoxanthinol
na
and amarouciaxanthin A were detected in plasma [8] and tissues, including the liver [9, 33, 34]. It may be due to the difference in fucoxanthin metabolism between humans and mice, as mice
ur
potentially metabolize and eliminate fucoxanthin faster than humans [31], which should be
Jo
considered when conducting studies for the effect of fucoxanthin in mice. In a pharmacokinetic study, fucoxanthinol and amarouciaxanthin A, but not fucoxanthin, were detected in the plasma, erythrocytes, liver, lung, kidney, heart, spleen, and adipose tissue when mice were orally administered with 160 nmol fucoxanthin [34]. After daily oral administration of fucoxanthin (160 nmol/day) for a week, fucoxanthin and its metabolites, i.e., fucoxanthinol and amarouciaxanthin A, were detected in tissue, but fucoxanthin was not present in the plasma. Consistently, after dietary fucoxanthin supplementation (0.128 ± 0.016 µmol/day) for 14 days, fucoxanthinol and amarouciaxanthin A were found in adipose tissues as well as in the plasma,
6
Journal Pre-proof liver, and kidney [35]. Therefore, fucoxanthin and its metabolites are delivered to different tissues to exert their biological effects with the metabolites being predominant forms [31, 34].
Safety and toxicity Studies have been conducted to assess the safety of fucoxanthin in vivo. A single oral administration of fucoxanthin at a dose of 1000 or 2000 mg/kg body weight did not elicit any
of
mortality and abnormality in ICR mice [12]. In addition, orally-administered fucoxanthin at a
ro
dose of 500 or 1000 mg/kg body weight for 30 days did not induce abnormal changes in tissues,
-p
including the liver, kidney, spleen, and gonadal tissues in ICR mice [12]. In rats administered
re
with a single oral dose of 2000 mg fucoxanthin/kg body weight or with 20 or 200 mg fucoxanthin/kg body weight for 13 weeks, no evident mortality or abnormalities were observed
lP
[13]. Therefore, it appears that fucoxanthin consumption is safe at least in the experimental
na
animal models.
The amount of fucoxanthin in brown seaweeds varies from 172 to 720 mg/kg dry weight [36],
ur
which is a relatively small amount compared with the doses of fucoxanthin administered in the
Jo
aforementioned animal studies. In addition, a dose of 2000 mg fucoxanthin/kg body weight in mice is equivalent to ~162 mg/kg body weight in humans, which may not be easily attainable by the consumption of seaweeds or dietary supplements in humans. Although the high amount of fucoxanthin seems to be safe in rodents, the safety investigation of fucoxanthin in humans is very limited. Ren et al. [37] administered human subjects with 5 mg of fucoxanthin/day for 5 weeks and the dose did not induce any adverse side effects. Fucoxanthin supplementation (1.6, 2.4, 4, or 8 mg) three times a day for 16 weeks showed no adverse effects in obese female subjects [38]. Two clinical trials are currently registered at the U. S. National Library of Medicine to determine
7
Journal Pre-proof the health benefits of daily 12 mg fucoxanthin consumption for 90 days (ClinicalTrials.gov Identifier: NCT03613740) [39] and daily 1650 mg fucoxanthin for up to 6 months (ClinicalTrials.gov Identifier: NCT02875392) [40]. More clinical trials are required to determine the safety and effective supplementation levels of fucoxanthin in humans.
Health benefits of fucoxanthin
of
Anti-inflammatory effect
ro
Inflammation is a biological process of the immune system in response to various
-p
external stimuli, including pathogens and toxic compounds, to counteract the inflammatory
re
insults [41]. Although inflammation is a crucial for the host defense, dysregulation of inflammatory responses leads to chronic inflammation, which is closely related to the
lP
pathogenesis of various diseases, including cardiovascular disease, diabetes, metabolic syndrome,
na
inflammatory bowel disease, rheumatoid arthritis, asthma, chronic obstructive lung disease, and cancer [42]. Therefore, it is essential to inhibit chronic inflammation for the prevention of the
ur
diseases mentioned above.
Jo
Studies have shown that fucoxanthin exerts anti-inflammatory action in vitro and in vivo. Fucoxanthin reduced the production of pro-inflammatory mediators, such as prostaglandin E2 (PGE2) and nitric oxide (NO), in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages [43, 44] and in LPS-treated BV-2 microglial cells [45]. This effect of fucoxanthin was attributed to its capacity to decrease protein levels of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS). In addition, the production of pro-inflammatory cytokines, such as interleukin1β (IL-1β), IL-6, and tumor necrosis factor α (TNFα), was significantly decreased by fucoxanthin in LPS-treated RAW 264.7 macrophages [43, 44] and in LPS-treated BV-2
8
Journal Pre-proof microglial cells with a concomitant decrease in cellular ROS levels [45]. The anti-inflammatory effect of fucoxanthin is likely due to its inhibitory effect on the activation of nuclear factor-κB (NF-κB) and the phosphorylation of mitogen-activated protein kinases, including c-JUN Nterminal kinase, extracellular signal-regulated kinase, and p38, in RAW 264.7 macrophages [46] and BV-2 microglial cells [45]. The anti-inflammatory properties of fucoxanthin have also been demonstrated in vivo.
of
Fucoxanthin supplementation at 0.6% by weight for 4 weeks significantly decreased serum
ro
levels of IL-1β and TNFα in mice fed a high-fat diet [47]. Also, dietary supplementation of
-p
fucoxanthin in a chow diet reduced the expression of TNFα and monocyte chemoattractant
re
protein-1 (MCP-1) in the white adipose tissue of KK-Ay mice [48]. In a mouse model of colitis, oral administration of fucoxanthin at a dose of 50 or 100 mg/kg body weight/day for 7 days
lP
ameliorated damages in the colon with reductions in PGE2 levels and COX-2 and NF-κB mRNA
na
and protein [49]. A recent study has shown that intragastric administration of fucoxanthin (200 mg/kg body weight) for 7 days before LPS injection to induce behavioral defects in mice
ur
improved depression and anxiety [50]. In this study, fucoxanthin reduced the protein level of
Jo
pro-inflammatory cytokines and enzymes, such as IL-1β, IL-6, and TNFα, iNOS, and COX-2, in the hippocampus, frontal cortex, and hypothalamus with a concomitant increase in phosphorylation of AMP-activated protein kinase (AMPK) and a decrease in NF-κB p65 protein. Furthermore, intravenous injection of fucoxanthin at a daily dose of 0.5 or 5 mg/kg body weight for 7 days improved ultraviolet B irradiation-induced corneal disorders in rats [51]. Importantly, protein levels of TNFα and vascular endothelial growth factor and infiltration of polymorphonuclear leukocytes in corneal tissues were decreased by fucoxanthin, suggesting that
9
Journal Pre-proof anti-inflammatory properties of fucoxanthin likely contribute to the prevention of corneal disorders.
Anti-cancer effect Cancer is the second leading cause of death, with an estimated 9.6 million deaths in 2018 worldwide [52]. Lung cancer is the most common cancer in both sexes, followed by breast
of
cancer in female, prostate cancer in male, and colorectal cancer [53]. Moreover, the most
ro
common cause of death related to cancer is also lung cancer, which is followed by colorectal,
-p
stomach, and liver cancer [53]. The diagnosis of cancer in the early stage is crucial to reduce
re
cancer-related death.
The preventive effect of fucoxanthin on cancer has been suggested in several cancer
lP
models. The anti-cancer effects of fucoxanthin are mediated by inducing cell cycle arrest and
na
apoptosis while inhibiting metastasis. Inhibiting the progression of the cell cycle is essential for cancer therapy [54]. Fucoxanthin is shown to reduce cell viability or proliferation by inducing
ur
cell cycle arrest in human gastric adenocarcinoma SGC-7901 and BGC-823 cells [55], human
Jo
non-small cell lung cancer cells, including A549 and H1299 [56], and the human bladder cancer T24 cell line [57]. The inhibitory effect of fucoxanthin on cell proliferation was mediated by increased p21, a cyclin-dependent kinase (CDK)-inhibitory protein, and decreased CDK-2, CDK-4, cyclin D1, and cyclin E in human bladder cancer T24 cell line. In addition, fucoxanthin can induce apoptosis of cells by increasing the expression of Bax while decreasing Bcl-2 expression in human cervical cancer SiHa cells [58], human gastric cancer cell line SGC7901 cells [59], human non-small cell lung cancer cells, including A549 and H1299 [56], human glioma cancer cell line U87 and U251 cells [60], and human bladder cancer
10
Journal Pre-proof cell line T24 cells [57]. Fucoxanthin also increased caspase-3 activity in these cancer cells. The effect of fucoxanthin on the induction of apoptotic cell death was mediated by inhibition of the phosphatidylinositol-3-kinase (PI3K)/ protein kinase B (Akt)/ mechanistic target of rapamycin pathway in cancer cells [60-62]. In addition, fucoxanthin induces apoptosis by reducing mRNA and protein expression of myeloid cell leukemia 1, an anti-apoptotic protein, signal transducer and activator of transcription 3 (STAT3), a transcription factor repressing apoptosis, and
of
phosphorylated active form of STAT3 in human gastric adenocarcinoma SGC-7901 or BGC-823
ro
cells [55]. In DLD-1 cells, fucoxanthin induces anoikis, which is apoptosis induced by cell
-p
detachment [63]. The effect of fucoxanthin on apoptosis in cancer cells was also demonstrated in
re
vivo. Fucoxanthin administration inhibited polyp formation and increased anoikis-like cells in colonic mucosa of mice treated with azoxymethane and dextran sodium sulfate for the induction
lP
of colorectal tumors [63].
na
The anti-cancer effect of fucoxanthin is also mediated by the inhibition of cancer cell migration or invasion. Fucoxanthin attenuated the migration of U87 and U251 cells in vitro,
ur
which was measured by the scratch wound healing assay and invasion of the cells through the
Jo
matrigel membrane using the trans-well assay [60]. In U87 and U251 cells, fucoxanthin decreased protein levels of matrix metalloproteinases 2 (MMP-2) and MMP-9 [60], which play a crucial role in tumor invasion and metastasis [64]. Fucoxanthin also reduced migration and invasion of highly metastatic murine B16-F10 melanoma cells, which was attributed to a reduction in the formation of actin fibers and the expression of MMP-9 in B16-F10 cells [65].
Anti-obesity effect
11
Journal Pre-proof Consumption of a diet high in fat and a lack of exercise cause an imbalance between energy intake and expenditure, leading to excessive accumulation of visceral fat. Obese individuals have high risks of developing type 2 diabetes, dyslipidemia, hypertension, cardiovascular disease, and other metabolic and inflammatory diseases [66, 67]. As these diseases increase mortality worldwide, it is essential to find strategies for preventing or treating obesity.
of
Studies have evaluated an anti-obesogenic effect of fucoxanthin. In db/db diabetic mice,
ro
fucoxanthin supplementation at 0.2% or 0.4% by weight for 6 weeks reduced plasma triglyceride
-p
and total cholesterol concentrations, which was attributed to the upregulation of hepatic protein
re
expression of peroxisome proliferator-activated receptor α (PPARα), phosphorylated acetylcoenzyme A carboxylase, and carnitine palmitoyltransferase 1 (CPT-1) [68]. Fucoxanthin also
lP
downregulated fatty acid synthase (FAS) protein via the activation of AMPK in the liver.
na
Moreover, in obese rats, daily administration of fucoxanthin (1 mg/kg body weight) for 8 weeks significantly improved serum lipid profiles, i.e., decreases in triglycerides, total cholesterol, and
ur
low-density lipoprotein cholesterol with increased high-density lipoprotein cholesterol [69]. The
Jo
changes in serum lipids were likely due to the upregulation of genes associated with energy expenditure and fatty acid oxidation, including PPARα and PPARγ coactivator 1α, in adipose tissue and the liver. Furthermore, when mice were fed a high-fat diet supplemented with fucoxanthin (0.05% or 0.2%, w/w) for 6 weeks, fucoxanthin significantly reduced fat-pad weight and adipocyte size [70]. This effect of fucoxanthin was attributed to the downregulation of mRNA and activity of lipogenic enzymes, including FAS, glucose-6-phosphate dehydrogenase (G6PD), and malic enzyme (ME) while upregulating genes involved in fatty acid β‐ oxidation. Gille et al. [71] also reported that consumption of fucoxanthin-rich extract (300 mg/kg body
12
Journal Pre-proof weight/day) from Phaeodactylum tribornutum for 26 days reduced body weight gain and white adipose tissue weight in mice fed a high-fat diet. Uncoupling protein 1 (UCP-1), a thermogenic mitochondrial protein, is usually expressed in brown adipose tissue. However, UCP-1 expression in white adipose tissue can be induced, leading to browning of white adipose tissue, consequently increasing thermogenesis [72, 73]. Studies have demonstrated that fucoxanthin stimulates UCP-1 expression in white adipose tissue.
of
Mice fed a fucoxanthin-rich fraction from edible seaweed, Undaria pinnatifida, showed a
ro
significant increase in UCP-1 expression in white adipose tissue concomitantly with decreased
-p
white adipose tissue weight [74]. Furthermore, consumption of AIN-93G diet containing fucoxanthin (0.2%, w/w) for 4 weeks significantly reduced white adipose tissue weight gain with
re
the induction of UCP-1 expression in diabetic/obese KK-Ay mice [75]. Also, when a high-fat diet
lP
containing 0.05% fucoxanthin (w/w) was given to mice for 6 weeks, there was a significant
na
increase in UCP-1 expression, which was likely to contribute to increased fatty acid β-oxidation and decreased fat production in white adipose tissue [70]. The expression of UCP-1 was
ur
increased in the subcutaneous white adipose tissues of mice fed a high-fat diet containing
Jo
fucoxanthin-rich extract [71]. Therefore, several studies have linked the anti-obesity effect of fucoxanthin to the induction of UCP-1 and beiging (or browing) of white adipose tissue. However, Rebello et al.[76] reported that fucoxanthin and fucoxanthinol did induce the browning of human adipocytes. Therefore, further studies are required to determine the effect of fucoxanthin on adipose tissue browning. Studies have also demonstrated that the anti-obesity effect of fucoxanthin is mediated, at least partly, by inhibiting adipocyte differentiation. Fucoxanthin significantly suppressed lipid accumulation during adipogenesis, which was mediated by inhibiting the expression of
13
Journal Pre-proof adipogenic and lipogenic factors, such as CCAAT/enhancer-binding protein alpha (C/EBPα), PPARγ, fatty acid-binding protein 4, diglyceride acyltransferase 1, and lysophosphatidic acid acyltransferase-θ in 3T3-L1 adipocytes [77]. The progression of adipocyte differentiation in 3T3L1 preadipocyte has three stages, i.e., early (days 0-2), intermediate (days 2-4), and late (days 47) [78]. Fucoxanthin showed different effects on adipocyte differentiation at each stage of differentiation. For instance, fucoxanthin promoted adipocyte differentiation at the early stage
of
while it suppressed at the later stage. At the early stage of adipocyte differentiation in 3T3-L1
ro
cells, fucoxanthin dose-dependently increased the protein levels of PPARγ, C/EBPα, and sterol
-p
regulatory element-binding protein 1c (SREBP-1c), while it inhibited the protein levels during
re
the intermediate and late stages of differentiation [78]. Furthermore, fucoxanthin and its metabolite, fucoxanthinol, showed an inhibitory effect on adipocyte differentiation in 3T3-L1
lP
adipocyte, as evidenced by reduced intracellular lipid accumulation and glycerol-3-phosphate
na
dehydrogenase (GPDH) activity, which was attributed to the downregulation of PPARγ expression [79]. Fucoxanthinol exhibited stronger inhibitory effects on adipocyte differentiation
ur
than fucoxanthin. Another metabolite of fucoxanthin, amarouciaxanthin A, also showed a
Jo
suppressive effect on 3T3-L1 adipocyte differentiation by decreasing the expression levels of transcription factors regulating adipocyte differentiation, e.g., PPAR and C/EBPα, and GPDH activity [80]. Furthermore, amarouciaxanthin A showed a stronger repressive effect on GPDH activity and the gene expression of fatty acid-binding protein 4, lipoprotein lipase, and glucose transporter 4 (GLUT4) than fucoxanthinol.
Anti-diabetic effect
14
Journal Pre-proof Diabetes mellitus is a complex metabolic disturbance closely related to obesity, which is characterized by increased blood glucose, mainly due to insulin resistance caused by excessive lipid accumulation in obesity [81, 82]. According to the World Health Organization, diabetes was the 7th leading cause of death in 2016 with the estimated number of death of 1.6 million [83]. The incidence of diabetes mellitus was increased from 108 million in 1980 to 422 million in 2014 [83]. Diabetes mellitus may cause severe complications, such as kidney disease,
of
blindness, heat attack, and stroke [84]. Therefore, it is vital to prevent or treat diabetes to avoid
ro
complications.
-p
Studies in mice have shown that fucoxanthin has an anti-diabetic effect by modulating
re
the insulin signaling pathway. In obese/diabetic KK-Ay mice, daily 0.1% fucoxanthin consumption for 27 days significantly reduced plasma glucose levels and glucose intolerance
lP
with concomitant decreases in the expression of pro-inflammatory genes, such as TNFα and
na
MCP-1, in white adipose tissue [85]. In the same mouse model, 0.2% fucoxanthin supplementation for 2 weeks decreased glucose and insulin levels in the circulation [86]. This
ur
anti-diabetic effect of fucoxanthin in KK-Ay mice was linked to enhanced GLUT4 translocation
Jo
to the cell membrane in soleus and extensor digitorum longus muscles, indicating insulin sensitivity was induced by fucoxanthin in mice. Indeed, fucoxanthin increased the expression of insulin receptor and the phosphorylation of Akt in soleus and extensor digitorum longus muscles [86]. Furthermore, KK-Ay mice fed 0.2% fucoxanthin (w/w) daily for 4 weeks showed significant reductions in leptin and TNFα mRNA levels in white adipose tissue, which contributed to the improvement of insulin resistance and reduction of blood glucose levels [75]. In db/db mice, fucoxanthin supplementation at 0.2% and 0.4% by weight reduced fasting blood glucose levels, improved intraperitoneal glucose tolerance, and enhanced intraperitoneal insulin
15
Journal Pre-proof tolerance [68]. The effect of fucoxanthin was mediated by insulin receptor substrate 1/ PI3K/Akt and AMPK signaling pathways in the liver and skeletal muscle. In a diet-induced obesity model, fucoxanthin markedly reduced the concentration of blood glucose and hemoglobin A1c levels with a concomitant decrease in plasma insulin and resistin concentrations [87]. Also, in high-fat diet-induced C57BL/6J mice, supplementation of fucoxanthin-rich wakame lipid extract markedly decreased plasma insulin and glucose levels concomitantly with an increase in GLUT4
of
expression and β3-adrenergic receptor but a decrease in MCP-1 expression [88]. As such,
ro
mounting evidence supports that fucoxanthin attenuates high-fat diet-induced insulin resistance
re
-p
in mouse models of obesity and diabetes.
Hepatoprotective effect
lP
Nonalcoholic fatty liver disease (NAFLD), the most common chronic liver disease in the
na
U.S., is characterized by excess lipid accumulation, inflammation, and fibrosis in the liver without significant alcohol consumption [89, 90]. Simple steatosis, i.e., excess lipid
ur
accumulation mainly in hepatocytes, is considered as benign, while the advanced form of
Jo
NAFLD such as nonalcoholic steatohepatitis has hepatocyte injury or cell death with inflammation and fibrosis in the liver [91]. It may further progress to cirrhosis with excess deposition of extracellular matrix. Studies have investigated the effect of fucoxanthin on the inhibition of hepatic lipid accumulation. In high-fat diet (20% fat, w/w)-fed C57BL/6J mice, the consumption of fucoxanthin at 0.05% or 0.2% level by weight for 6 weeks significantly reduced hepatic triglyceride and cholesterol accumulation [87]. This hepatoprotective effect of fucoxanthin was likely mediated by suppression of hepatic lipogenesis with increased hepatic lipolysis. The
16
Journal Pre-proof activities of enzymes related to fatty acid synthesis, including FAS, G6PD, and ME, were significantly suppressed by fucoxanthin, while genes related to fatty acid β-oxidation, e.g., acylCoA oxidase1 and PPARα, were significantly induced by fucoxanthin in the liver [87]. Also, supplementation of fucoxanthin-rich extract of Undaria pinnatifida, an edible brown alga, significantly reduced hepatic lipid content in high-fat diet-induced obesity mice, which was attributed to reduced activities of enzymes involved in fatty acid synthesis, including FAS, ME,
of
and G6PD, with a concomitant increase in fatty acid oxidation in the liver [92]. Also, the
ro
activities of enzymes for cholesterol metabolism, such as 3-hydroxy-3-methylglutaryl coenzyme
-p
A reductase and acyl-CoA cholesterol acyltransferase, were significantly decreased by the supplementation of fucoxanthin-rich seaweed extract [92].
re
It has been shown that fucoxanthin can alter lipid composition in the liver. In KK-Ay mice,
lP
fucoxanthin downregulated the expression of hepatic stearoyl-coenzyme A desaturase-1 [93],
na
which catalyzes the biosynthesis of monounsaturated fatty acids from saturated fatty acids and improves insulin sensitivity in diet-induced obesity mice [94]. Therefore, fucoxanthin feeding
ur
changed fatty acid composition such as stearic acid and oleic acid in the liver of KK-Ay mice,
Jo
decreasing the ratios of oleic acid to stearic acid. Interestingly, oleic acid-induced hepatic lipid accumulation and lipid peroxidation were significantly attenuated by fucoxanthin in FL83B hepatocytes [95]. The inhibitory effect of fucoxanthin on lipid accumulation was attributed to the suppression of lipogenic genes, such as SREBP-1c, PPARγ and FAS, while increasing the expression of genes for lipolysis and β-oxidation, e.g., adipose triglyceride lipase, phosphorylated hormone-sensitive lipase, CPT-1, CPT-2, and PPARα. The beneficial effect of fucoxanthin on the prevention of NAFLD may also be due to its anti-fibrogenic properties. The activation of hepatic stellate cells (HSCs) is an essential event in
17
Journal Pre-proof the development of liver fibrosis. Upon liver injury, quiescent HSCs are transdifferentiated into activated HSCs, which have high expression of fibrogenic genes including, α smooth muscle actin and procollagen type I a1 [96]. We recently demonstrated that fucoxanthin attenuated transforming growth factor β1 (TGFβ1)-induced pro-fibrogenic gene expression by the inhibition of SMA- and MAD-related protein activation in LX-2 cells as well as primary human HSCs [96]. Also, using primary mouse HSCs, we showed that fucoxanthin inhibited the transdifferentiation
of
of quiescent HSCs to activated HSCs in the same study. Furthermore, fucoxanthin prevented
ro
TGFβ1-induced ROS accumulation, at least in part, by inhibiting the expression of NADPH
-p
oxidase 4, an enzyme that generates ROS, which is likely to contribute to the anti-fibrogenic
re
effect of fucoxanthin.
Studies have shown a protective effect of fucoxanthin on oxidative stress in hepatic cell
lP
lines. Ferric nitrilotriacetate (Fe-NTA) treatment significantly reduced cell viability, while the
na
presence of fucoxanthin significantly attenuated Fe-NTA-inhibited cell proliferation in hepatic BNL CL.2 cells [97]. This protective effect of fucoxanthin is likely attributed to decreased
ur
intracellular ROS, thiobarbituric acid reactive substances level, and protein carbonyl contents
Jo
with increased glutathione levels [97]. Also in HepG2 cells, fucoxanthin prevented arachidonic acid+ iron-induced oxidative stress and increased expression of apoptosis-related proteins, mitochondrial dysfunction, and autophagy, which were attributed to the liver kinase B1-AMPKα signaling pathway [98].
Conclusion Fucoxanthin, a xanthophyll carotenoid, has a unique chemical structure that confers a strong antioxidant capacity by scavenging singlet molecular oxygen and free radicals. Studies
18
Journal Pre-proof have demonstrated the potential health benefits of fucoxanthin, including anti-inflammatory, anti-cancer, anti-obesity, anti-diabetic, and hepatoprotective effects (Figure 3). Therefore, fucoxanthin has potential as a preventative/therapeutic agent for the diseases. However, although no toxic effect of fucoxanthin has been reported from animal studies, clinical trials to determine the safety of fucoxanthin consumption in humans are needed. Furthermore, as fucoxanthin can be metabolized to fucoxanthinol in the gastrointestinal tract and further to amarouciaxanthin A in
of
the liver, in vitro studies using the fucoxanthin metabolites are necessary for the better
-p
ro
mechanistic understanding of how fucoxanthin exerts its health benefits.
re
Acknowledgments
This work was supported by USDA Multistate Hatch CONS00992 to J.-Y. Lee and
Jo
ur
na
lP
USDA Hatch CONS00978 to Y.-K Park.
19
Journal Pre-proof References [1] J. Peng, J.P. Yuan, C.F. Wu, J.H. Wang, Fucoxanthin, a marine carotenoid present in brown seaweeds and diatoms: metabolism and bioactivities relevant to human health, Marine drugs, 9 (2011) 1806-1828. [2] R.K. Sangeetha, N. Bhaskar, S. Divakar, V. Baskaran, Bioavailability and metabolism of fucoxanthin in rats: structural characterization of metabolites by LC-MS (APCI), Molecular and cellular biochemistry, 333 (2010) 299-310.
of
[3] S.M. Kim, Y.J. Jung, O.N. Kwon, K.H. Cha, B.H. Um, D. Chung, C.H. Pan, A potential commercial source of fucoxanthin extracted from the microalga Phaeodactylum tricornutum, Appl Biochem Biotechnol, 166 (2012) 1843-1855.
ro
[4] L.J. Wang, Y. Fan, R.L. Parsons, G.R. Hu, P.Y. Zhang, F.L. Li, A Rapid Method for the Determination of Fucoxanthin in Diatom, Marine drugs, 16 (2018).
re
-p
[5] N.M. Sachindra, E. Sato, H. Maeda, M. Hosokawa, Y. Niwano, M. Kohno, K. Miyashita, Radical scavenging and singlet oxygen quenching activity of marine carotenoid fucoxanthin and its metabolites, Journal of agricultural and food chemistry, 55 (2007) 8516-8522.
lP
[6] S. Xia, K. Wang, L. Wan, A. Li, Q. Hu, C. Zhang, Production, characterization, and antioxidant activity of fucoxanthin from the marine diatom Odontella aurita, Marine drugs, 11 (2013) 2667-2681.
na
[7] D. Zhao, S.-M. Kim, C.-H. Pan, D. Chung, Effects of heating, aerial exposure and illumination on stability of fucoxanthin in canola oil, Food chemistry, 145 (2014) 505-513.
ur
[8] T. Sugawara, V. Baskaran, W. Tsuzuki, A. Nagao, Brown algae fucoxanthin is hydrolyzed to fucoxanthinol during absorption by Caco-2 human intestinal cells and mice, The Journal of nutrition, 132 (2002) 946-951.
Jo
[9] A. Asai, T. Sugawara, H. Ono, A. Nagao, Biotransformation of fucoxanthinol into amarouciaxanthin A in mice and HepG2 cells: formation and cytotoxicity of fucoxanthin metabolites, Drug Metab Dispos, 32 (2004) 205-211. [10] T. Sugawara, K. Yamashita, A. Asai, A. Nagao, T. Shiraishi, I. Imai, T. Hirata, Esterification of xanthophylls by human intestinal Caco-2 cells, Archives of biochemistry and biophysics, 483 (2009) 205-212. [11] K. Miyashita, S. Nishikawa, F. Beppu, T. Tsukui, M. Abe, M. Hosokawa, The allenic carotenoid fucoxanthin, a novel marine nutraceutical from brown seaweeds, Journal of the science of food and agriculture, 91 (2011) 1166-1174. [12] F. Beppu, Y. Niwano, T. Tsukui, M. Hosokawa, K. Miyashita, Single and repeated oral dose toxicity study of fucoxanthin (FX), a marine carotenoid, in mice, J Toxicol Sci, 34 (2009) 501510.
20
Journal Pre-proof [13] K. Iio, Y. Okada, M. Ishikura, [Single and 13-week oral toxicity study of fucoxanthin oil from microalgae in rats], Shokuhin Eiseigaku Zasshi, 52 (2011) 183-189. [14] Letter from FDA CFSAN to Algatechnologies regarding NDI 1048 – Fucoxanthin. [15] A. Agócs, E. Murillo, E. Turcsi, S. Béni, A. Darcsi, Á. Szappanos, T. Kurtán, J. Deli, Isolation of allene carotenoids from mamey, Journal of Food Composition and Analysis, 65 (2018) 1-5. [16] D. Pádua, E. Rocha, D. Gargiulo, A.A. Ramos, Bioactive compounds from brown seaweeds: Phloroglucinol, fucoxanthin and fucoidan as promising therapeutic agents against breast cancer, Phytochemistry letters, 2015 v.14 (2015) pp. 91-98.
ro
of
[17] W. Stahl, H. Sies, Antioxidant activity of carotenoids, Molecular aspects of medicine, 24 (2003) 345-351.
-p
[18] F. Ramel, S. Birtic, S. Cuine, C. Triantaphylides, J.L. Ravanat, M. Havaux, Chemical quenching of singlet oxygen by carotenoids in plants, Plant physiology, 158 (2012) 1267-1278.
re
[19] P.F. Conn, W. Schalch, T.G. Truscott, The singlet oxygen and carotenoid interaction, Journal of photochemistry and photobiology. B, Biology, 11 (1991) 41-47.
lP
[20] N.J. Miller, J. Sampson, L.P. Candeias, P.M. Bramley, C.A. Rice-Evans, Antioxidant activities of carotenes and xanthophylls, FEBS letters, 384 (1996) 240-242.
na
[21] C.L. Liu, A.L. Liang, M.L. Hu, Protective effects of fucoxanthin against ferric nitrilotriacetate-induced oxidative stress in murine hepatic BNL CL.2 cells, Toxicol In Vitro, 25 (2011) 1314-1319.
Jo
ur
[22] J. Zeng, Y. Zhang, J. Ruan, Z. Yang, C. Wang, Z. Hong, Z. Zuo, Protective effects of fucoxanthin and fucoxanthinol against tributyltin-induced oxidative stress in HepG2 cells, Environ Sci Pollut Res Int, 25 (2018) 5582-5589. [23] J. Zheng, M.J. Piao, K.C. Kim, C.W. Yao, J.W. Cha, J.W. Hyun, Fucoxanthin enhances the level of reduced glutathione via the Nrf2-mediated pathway in human keratinocytes, Marine drugs, 12 (2014) 4214-4230. [24] C.L. Liu, Y.T. Chiu, M.L. Hu, Fucoxanthin enhances HO-1 and NQO1 expression in murine hepatic BNL CL.2 cells through activation of the Nrf2/ARE system partially by its prooxidant activity, Journal of agricultural and food chemistry, 59 (2011) 11344-11351. [25] V. Shete, L. Quadro, Mammalian metabolism of beta-carotene: gaps in knowledge, Nutrients, 5 (2013) 4849-4868. [26] E. Reboul, Absorption of vitamin A and carotenoids by the enterocyte: focus on transport proteins, Nutrients, 5 (2013) 3563-3581.
21
Journal Pre-proof [27] E.H. Harrison, Mechanisms involved in the intestinal absorption of dietary vitamin A and provitamin A carotenoids, Biochimica et biophysica acta, 1821 (2012) 70-77. [28] C. Kiefer, E. Sumser, M.F. Wernet, J. Von Lintig, A class B scavenger receptor mediates the cellular uptake of carotenoids in Drosophila, Proceedings of the National Academy of Sciences of the United States of America, 99 (2002) 10581-10586. [29] A. Eroglu, E.H. Harrison, Carotenoid metabolism in mammals, including man: formation, occurrence, and function of apocarotenoids, Journal of lipid research, 54 (2013) 1719-1730.
of
[30] Y. Zhang, H. Wu, H. Wen, H. Fang, Z. Hong, R. Yi, R. Liu, Simultaneous Determination of Fucoxanthin and Its Deacetylated Metabolite Fucoxanthinol in Rat Plasma by Liquid Chromatography-Tandem Mass Spectrometry, Marine drugs, 13 (2015) 6521-6536.
ro
[31] T. Hashimoto, Y. Ozaki, M. Mizuno, M. Yoshida, Y. Nishitani, T. Azuma, A. Komoto, T. Maoka, Y. Tanino, K. Kanazawa, Pharmacokinetics of fucoxanthinol in human plasma after the oral administration of kombu extract, The British journal of nutrition, 107 (2012) 1566-1569.
re
-p
[32] A. Asai, L. Yonekura, A. Nagao, Low bioavailability of dietary epoxyxanthophylls in humans, The British journal of nutrition, 100 (2008) 273-277.
lP
[33] T. Tsukui, N. Baba, M. Hosokawa, T. Sashima, K. Miyashita, Enhancement of hepatic docosahexaenoic acid and arachidonic acid contents in C57BL/6J mice by dietary fucoxanthin, Fisheries Science, 75 (2009) 261-263.
na
[34] T. Hashimoto, Y. Ozaki, M. Taminato, S.K. Das, M. Mizuno, K. Yoshimura, T. Maoka, K. Kanazawa, The distribution and accumulation of fucoxanthin and its metabolites after oral administration in mice, The British journal of nutrition, 102 (2009) 242-248.
Jo
ur
[35] L. Yonekura, M. Kobayashi, M. Terasaki, A. Nagao, Keto-carotenoids are the major metabolites of dietary lutein and fucoxanthin in mouse tissues, The Journal of nutrition, 140 (2010) 1824-1831. [36] H. Dominguez, Functional ingredients from algae for foods and nutraceuticals, Elsevier, 2013. [37] R. Ren, Y. Azuma, T. Ojima, T. Hashimoto, M. Mizuno, Y. Nishitani, M. Yoshida, T. Azuma, K. Kanazawa, Modulation of platelet aggregation-related eicosanoid production by dietary F-fucoidan from brown alga Laminaria japonica in human subjects, The British journal of nutrition, 110 (2013) 880-890. [38] M. Abidov, Z. Ramazanov, R. Seifulla, S. Grachev, The effects of Xanthigen in the weight management of obese premenopausal women with non-alcoholic fatty liver disease and normal liver fat, Diabetes Obes Metab, 12 (2010) 72-81. [39] Effect of Fucoxanthin on the Components of the Metabolic Syndrome, Insulin Sensitivity and Insulin Secretion.
22
Journal Pre-proof [40] Fucoidan Improves the Metabolic Profiles of Patients With Non-alcoholic Fatty Liver Disease (NAFLD). [41] L. Chen, H. Deng, H. Cui, J. Fang, Z. Zuo, J. Deng, Y. Li, X. Wang, L. Zhao, Inflammatory responses and inflammation-associated diseases in organs, Oncotarget, 9 (2018) 7204-7218. [42] J. Zhong, G. Shi, Editorial: Regulation of Inflammation in Chronic Disease, Front Immunol, 10 (2019) 737.
of
[43] S.J. Heo, W.J. Yoon, K.N. Kim, G.N. Ahn, S.M. Kang, D.H. Kang, A. Affan, C. Oh, W.K. Jung, Y.J. Jeon, Evaluation of anti-inflammatory effect of fucoxanthin isolated from brown algae in lipopolysaccharide-stimulated RAW 264.7 macrophages, Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association, 48 (2010) 2045-2051.
-p
ro
[44] K. Shiratori, K. Ohgami, I. Ilieva, X.H. Jin, Y. Koyama, K. Miyashita, K. Yoshida, S. Kase, S. Ohno, Effects of fucoxanthin on lipopolysaccharide-induced inflammation in vitro and in vivo, Exp Eye Res, 81 (2005) 422-428.
lP
re
[45] D. Zhao, S.H. Kwon, Y.S. Chun, M.Y. Gu, H.O. Yang, Anti-Neuroinflammatory Effects of Fucoxanthin via Inhibition of Akt/NF-kappaB and MAPKs/AP-1 Pathways and Activation of PKA/CREB Pathway in Lipopolysaccharide-Activated BV-2 Microglial Cells, Neurochemical research, 42 (2017) 667-677.
na
[46] K.N. Kim, S.J. Heo, W.J. Yoon, S.M. Kang, G. Ahn, T.H. Yi, Y.J. Jeon, Fucoxanthin inhibits the inflammatory response by suppressing the activation of NF-kappaB and MAPKs in lipopolysaccharide-induced RAW 264.7 macrophages, European journal of pharmacology, 649 (2010) 369-375.
Jo
ur
[47] C.P. Tan, Y.H. Hou, First evidence for the anti-inflammatory activity of fucoxanthin in high-fat-diet-induced obesity in mice and the antioxidant functions in PC12 cells, Inflammation, 37 (2014) 443-450. [48] H. Maeda, S. Kanno, M. Kodate, M. Hosokawa, K. Miyashita, Fucoxanthinol, Metabolite of Fucoxanthin, Improves Obesity-Induced Inflammation in Adipocyte Cells, Marine drugs, 13 (2015) 4799-4813. [49] Y.P. Yang, Q.Y. Tong, S.H. Zheng, M.D. Zhou, Y.M. Zeng, T.T. Zhou, Anti-inflammatory effect of fucoxanthin on dextran sulfate sodium-induced colitis in mice, Nat Prod Res, (2018) 1-5. [50] X. Jiang, G. Wang, Q. Lin, Z. Tang, Q. Yan, X. Yu, Fucoxanthin prevents lipopolysaccharide-induced depressive-like behavior in mice via AMPK- NF-kappaB pathway, Metab Brain Dis, 34 (2019) 431-442. [51] S.J. Chen, C.J. Lee, T.B. Lin, H.J. Liu, S.Y. Huang, J.Z. Chen, K.W. Tseng, Inhibition of Ultraviolet B-Induced Expression of the Proinflammatory Cytokines TNF-alpha and VEGF in the Cornea by Fucoxanthin Treatment in a Rat Model, Marine drugs, 14 (2016) 13.
23
Journal Pre-proof [52] Cancer, World Health Organization, in, 2018. [53] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA: a cancer journal for clinicians, 68 (2018) 394-424. [54] C.C. Mills, E.A. Kolb, V.B. Sampson, Development of Chemotherapy with Cell-Cycle Inhibitors for Adult and Pediatric Cancer Therapy, Cancer Res, 78 (2018) 320-325. [55] R.X. Yu, R.T. Yu, Z. Liu, Inhibition of two gastric cancer cell lines induced by fucoxanthin involves downregulation of Mcl-1 and STAT3, Hum Cell, 31 (2018) 50-63.
of
[56] C. Mei, S. Zhou, L. Zhu, J. Ming, F. Zeng, R. Xu, Antitumor Effects of Laminaria Extract Fucoxanthin on Lung Cancer, Marine drugs, 15 (2017).
-p
ro
[57] L. Wang, Y. Zeng, Y. Liu, X. Hu, S. Li, Y. Wang, L. Li, Z. Lei, Z. Zhang, Fucoxanthin induces growth arrest and apoptosis in human bladder cancer T24 cells by up-regulation of p21 and down-regulation of mortalin, Acta Biochim Biophys Sin (Shanghai), 46 (2014) 877-884.
lP
re
[58] Y. Jin, S. Qiu, N. Shao, J. Zheng, Fucoxanthin and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Synergistically Promotes Apoptosis of Human Cervical Cancer Cells by Targeting PI3K/Akt/NF-kappaB Signaling Pathway, Medical science monitor : international medical journal of experimental and clinical research, 24 (2018) 11-18.
na
[59] Y. Zhu, J. Cheng, Z. Min, T. Yin, R. Zhang, W. Zhang, L. Hu, Z. Cui, C. Gao, S. Xu, C. Zhang, X. Hu, Effects of fucoxanthin on autophagy and apoptosis in SGC-7901cells and the mechanism, Journal of cellular biochemistry, 119 (2018) 7274-7284.
Jo
ur
[60] Y. Liu, J. Zheng, Y. Zhang, Z. Wang, Y. Yang, M. Bai, Y. Dai, Fucoxanthin Activates Apoptosis via Inhibition of PI3K/Akt/mTOR Pathway and Suppresses Invasion and Migration by Restriction of p38-MMP-2/9 Pathway in Human Glioblastoma Cells, Neurochemical research, 41 (2016) 2728-2751. [61] J. Yang, C. Pi, G. Wang, Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 103 (2018) 699-707. [62] C. Xu, Q. Wang, X. Feng, Y. Bo, Effect of evodiagenine mediates photocytotoxicity on human breast cancer cells MDA-MB-231 through inhibition of PI3K/AKT/mTOR and activation of p38 pathways, Fitoterapia, 99 (2014) 292-299. [63] M. Terasaki, T. Iida, F. Kikuchi, K. Tamura, T. Endo, Y. Kuramitsu, T. Tanaka, H. Maeda, K. Miyashita, M. Mutoh, Fucoxanthin potentiates anoikis in colon mucosa and prevents carcinogenesis in AOM/DSS model mice, The Journal of nutritional biochemistry, 64 (2019) 198-205. [64] K. Nabeshima, T. Inoue, Y. Shimao, T. Sameshima, Matrix metalloproteinases in tumor invasion: role for cell migration, Pathology international, 52 (2002) 255-264. 24
Journal Pre-proof [65] T.W. Chung, H.J. Choi, J.Y. Lee, H.S. Jeong, C.H. Kim, M. Joo, J.Y. Choi, C.W. Han, S.Y. Kim, J.S. Choi, K.T. Ha, Marine algal fucoxanthin inhibits the metastatic potential of cancer cells, Biochemical and biophysical research communications, 439 (2013) 580-585. [66] C. Torres-Fuentes, H. Schellekens, T.G. Dinan, J.F. Cryan, A natural solution for obesity: bioactives for the prevention and treatment of weight gain. A review, Nutritional neuroscience, 18 (2015) 49-65. [67] M.A. Gammone, N. D'Orazio, Anti-obesity activity of the marine carotenoid fucoxanthin, Marine drugs, 13 (2015) 2196-2214.
of
[68] Y. Zhang, W. Xu, X. Huang, Y. Zhao, Q. Ren, Z. Hong, M. Huang, X. Xing, Fucoxanthin ameliorates hyperglycemia, hyperlipidemia and insulin resistance in diabetic mice partially through irs-1/pi3k/akt and ampk pathways, Journal of functional foods, 48 (2018) 515-524.
-p
ro
[69] A. Grasa-López, Á. Miliar-García, L. Quevedo-Corona, N. Paniagua-Castro, G. EscalonaCardoso, E. Reyes-Maldonado, M.-E. Jaramillo-Flores, Undaria pinnatifida and fucoxanthin ameliorate lipogenesis and markers of both inflammation and cardiovascular dysfunction in an animal model of diet-induced obesity, Marine drugs, 14 (2016) 148.
lP
re
[70] M.N. Woo, S.M. Jeon, Y.C. Shin, M.K. Lee, M.A. Kang, M.S. Choi, Anti‐ obese property of fucoxanthin is partly mediated by altering lipid‐ regulating enzymes and uncoupling proteins of visceral adipose tissue in mice, Molecular nutrition & food research, 53 (2009) 1603-1611.
na
[71] A. Gille, B. Stojnic, F. Derwenskus, A. Trautmann, U. Schmid-Staiger, C. Posten, K. Briviba, A. Palou, M.L. Bonet, J. Ribot, A Lipophilic Fucoxanthin-Rich Phaeodactylum tricornutum Extract Ameliorates Effects of Diet-Induced Obesity in C57BL/6J Mice, Nutrients, 11 (2019).
Jo
ur
[72] T. Yoneshiro, S. Aita, M. Matsushita, T. Kayahara, T. Kameya, Y. Kawai, T. Iwanaga, M. Saito, Recruited brown adipose tissue as an antiobesity agent in humans, The Journal of clinical investigation, 123 (2013) 3404-3408. [73] J. Wu, P. Cohen, B.M. Spiegelman, Adaptive thermogenesis in adipocytes: is beige the new brown?, Genes & development, 27 (2013) 234-250. [74] H. Maeda, M. Hosokawa, T. Sashima, K. Funayama, K. Miyashita, Fucoxanthin from edible seaweed, Undaria pinnatifida, shows antiobesity effect through UCP1 expression in white adipose tissues, Biochemical and biophysical research communications, 332 (2005) 392-397. [75] H. Maeda, M. Hosokawa, T. Sashima, K. Miyashita, Dietary combination of fucoxanthin and fish oil attenuates the weight gain of white adipose tissue and decreases blood glucose in obese/diabetic KK-Ay mice, J. of Agricultural and Food Chemistry, 55 (2007) 7701-7706. [76] C.J. Rebello, F.L. Greenway, W.D. Johnson, D. Ribnicky, A. Poulev, K. Stadler, A.A. Coulter, Fucoxanthin and Its Metabolite Fucoxanthinol Do Not Induce Browning in Human Adipocytes, Journal of agricultural and food chemistry, 65 (2017) 10915-10924.
25
Journal Pre-proof [77] M.J. Seo, Y.J. Seo, C.H. Pan, O.H. Lee, K.J. Kim, B.Y. Lee, Fucoxanthin suppresses lipid accumulation and ROS production during differentiation in 3T3‐ L1 adipocytes, Phytotherapy research, 30 (2016) 1802-1808. [78] S.-I. Kang, H.-C. Ko, H.-S. Shin, H.-M. Kim, Y.-S. Hong, N.-H. Lee, S.-J. Kim, Fucoxanthin exerts differing effects on 3T3-L1 cells according to differentiation stage and inhibits glucose uptake in mature adipocytes, Biochemical and biophysical research communications, 409 (2011) 769-774. [79] H. Maeda, M. Hosokawa, T. Sashima, N. Takahashi, T. Kawada, K. Miyashita, Fucoxanthin and its metabolite, fucoxanthinol, suppress adipocyte differentiation in 3T3-L1 cells, International journal of molecular medicine, 18 (2006) 147-152.
-p
ro
of
[80] M.-J. Yim, M. Hosokawa, Y. Mizushina, H. Yoshida, Y. Saito, K. Miyashita, Suppressive effects of amarouciaxanthin A on 3T3-L1 adipocyte differentiation through down-regulation of PPARγ and C/EBPα mRNA expression, Journal of agricultural and food chemistry, 59 (2011) 1646-1652.
re
[81] H. Zhang, Y. Tang, Y. Zhang, S. Zhang, J. Qu, X. Wang, R. Kong, C. Han, Z. Liu, Fucoxanthin: A promising medicinal and nutritional ingredient, Evidence-based complementary and alternative medicine, 2015 (2015).
lP
[82] A. Kawee-Ai, A.T. Kim, S.M. Kim, Inhibitory activities of microalgal fucoxanthin against α-amylase, α-glucosidase, and glucose oxidase in 3T3-L1 cells linked to type 2 diabetes, Journal of Oceanology and Limnology, 37 (2019) 928-937.
na
[83] Diabetes, in, World Health Organization, 2018.
ur
[84] J.M. Forbes, M.E. Cooper, Mechanisms of diabetic complications, Physiological reviews, 93 (2013) 137-188.
Jo
[85] H. Maeda, S. Kanno, M. Kodate, M. Hosokawa, K. Miyashita, Fucoxanthinol, metabolite of fucoxanthin, improves obesity-induced inflammation in adipocyte cells, Marine drugs, 13 (2015) 4799-4813. [86] S. Nishikawa, M. Hosokawa, K. Miyashita, Fucoxanthin promotes translocation and induction of glucose transporter 4 in skeletal muscles of diabetic/obese KK-Ay mice, Phytomedicine, 19 (2012) 389-394. [87] M.-N. Woo, S.-M. Jeon, H.-J. Kim, M.-K. Lee, S.-K. Shin, Y.C. Shin, Y.-B. Park, M.-S. Choi, Fucoxanthin supplementation improves plasma and hepatic lipid metabolism and blood glucose concentration in high-fat fed C57BL/6N mice, Chemico-Biological Interactions, 186 (2010) 316-322. [88] H. Maeda, M. Hosokawa, T. Sashima, K. Murakami-Funayama, K. Miyashita, Anti-obesity and anti-diabetic effects of fucoxanthin on diet-induced obesity conditions in a murine model, Molecular Medicine Reports, 2 (2009) 897-902.
26
Journal Pre-proof [89] C. Dorn, M.-O. Riener, G. Kirovski, M. Saugspier, K. Steib, T.S. Weiss, E. Gäbele, G. Kristiansen, A. Hartmann, C. Hellerbrand, Expression of fatty acid synthase in nonalcoholic fatty liver disease, International journal of clinical and experimental pathology, 3 (2010) 505. [90] N. Chalasani, Z. Younossi, J.E. Lavine, A.M. Diehl, E.M. Brunt, K. Cusi, M. Charlton, A.J. Sanyal, The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association, Hepatology, 55 (2012) 2005-2023.
of
[91] E.M. Brunt, V.W. Wong, V. Nobili, C.P. Day, S. Sookoian, J.J. Maher, E. Bugianesi, C.B. Sirlin, B.A. Neuschwander-Tetri, M.E. Rinella, Nonalcoholic fatty liver disease, Nature reviews. Disease primers, 1 (2015) 15080.
-p
ro
[92] S.M. Jeon, H.J. Kim, M.N. Woo, M.K. Lee, Y.C. Shin, Y.B. Park, M.S. Choi, Fucoxanthin‐ rich seaweed extract suppresses body weight gain and improves lipid metabolism in high‐ fat‐ fed C57BL/6J mice, Biotechnology journal, 5 (2010) 961-969.
re
[93] F. Beppu, M. Hosokawa, M.J. Yim, T. Shinoda, K. Miyashita, Down‐ regulation of hepatic stearoyl‐ CoA desaturase‐ 1 expression by Fucoxanthin via leptin signaling in diabetic/obese KK‐ Ay mice, Lipids, 48 (2013) 449-455.
lP
[94] J.M. Ntambi, M. Miyazaki, J.P. Stoehr, H. Lan, C.M. Kendziorski, B.S. Yandell, Y. Song, P. Cohen, J.M. Friedman, A.D. Attie, Loss of stearoyl–CoA desaturase-1 function protects mice against adiposity, Proceedings of the National Academy of Sciences, 99 (2002) 11482-11486.
ur
na
[95] Y.-H. Chang, Y.-L. Chen, W.-C. Huang, C.-J. Liou, Fucoxanthin attenuates fatty acidinduced lipid accumulation in FL83B hepatocytes through regulated Sirt1/AMPK signaling pathway, Biochemical and biophysical research communications, 495 (2018) 197-203.
Jo
[96] M.-B. Kim, M. Bae, S. Hu, H. Kang, Y.-K. Park, J.-Y. Lee, Fucoxanthin exerts antifibrogenic effects in hepatic stellate cells, Biochemical and biophysical research communications, (2019). [97] C.-L. Liu, A.-L. Liang, M.-L. Hu, Protective effects of fucoxanthin against ferric nitrilotriacetate-induced oxidative stress in murine hepatic BNL CL. 2 cells, Toxicology in Vitro, 25 (2011) 1314-1319. [98] E.J. Jang, S.C. Kim, J.-H. Lee, J.R. Lee, I.K. Kim, S.Y. Baek, Y.W. Kim, Fucoxanthin, the constituent of Laminaria japonica, triggers AMPK-mediated cytoprotection and autophagy in hepatocytes under oxidative stress, BMC complementary and alternative medicine, 18 (2018) 97.
27
Journal Pre-proof Figure legends Figure 1. Structure of fucoxanthin.
Figure 2. Biotransformation of fucoxanthin. Fucoxanthin can be hydrolyzed to fucoxanthinol in the gastrointestinal tract and further converted into amarouciaxanthin A in the liver
of
Figure 3. Health benefits of fucoxanthin and its metabolites. Fucoxanthin and its metabolites,
ro
fucoxanthinol and amarouciaxanthin A, exert anti-inflammatory, anti-cancer, anti-obesity, anti-
Jo
ur
na
lP
re
-p
diabetic, and hepatoprotective effects.
28
Journal Pre-proof Declaration of interests
☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Jo
ur
na
lP
re
-p
ro
of
☐The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:
29
Figure 1
Figure 2
Figure 3