Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Nutr Cancer ; 74(10): 3651-3661, 2022.
Article in English | MEDLINE | ID: mdl-35695489

ABSTRACT

Fucoxanthin (Fx) is a critical pigment required for photosynthesis in brown algae and microalgae. Fx is also a dietary marine carotenoid that with potent anticancer activity in vitro and in vivo. Some popular light meals for increased satiety, such as biscuits, cereals, and crackers, are frequently fortified with micronutrients for human health benefits. However, data on the anticancer potential of Fx-supplemented light meals in humans and animal models remain limited. In the present study, we investigated the anticancer effects of a Fx-supplemented biscuit using a carcinogenic murine azoxymethane/dextran sodium sulfate (AOM/DSS) model. We observed that periodic administration of biscuits containing 0.3% Fx (Fx-biscuit) at an interval of 3 days (each 15 h) per week for 15 weeks significantly inhibited colorectal carcinogenesis in AOM/DSS mice. Comprehensive gene analysis demonstrated that the Fx-biscuit significantly altered the expression of 138 genes in the colorectal mucosal tissue of the mice. In particular, the expression of heat shock protein 70 (HSP70) genes, Hspa1b (-35.7-fold) and Hspa1a (-34.9-fold), was markedly downregulated. HSP70 is a polyfunctional chaperone protein that is involved in cancer development. Compared to the control-biscuit group, the number of cells with markedly high fluorescence for HSP70 protein (HSP70high) in colorectal mucosal crypts and adenocarcinomas significantly reduced by 0.3- and 0.2-fold, respectively, in the Fx-biscuit group. Our results suggested that Fx-biscuit possesses chemopreventive potential in the colorectal cancer of AOM/DSS mice via the downregulation of HSP70.


Subject(s)
Colitis , Colorectal Neoplasms , Animals , Azoxymethane/toxicity , Carcinogenesis , Colitis/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Dextran Sulfate/toxicity , Disease Models, Animal , Humans , Mice , Xanthophylls
2.
Nutr Cancer ; 74(1): 357-371, 2022.
Article in English | MEDLINE | ID: mdl-33590779

ABSTRACT

Fucoxanthin and its metabolite fucoxanthinol (FxOH), highly polar xanthophylls, exert strong anticancer effects against many cancer cell types. However, the effects of Fx and FxOH on pancreatic cancer, a high mortality cancer, remain unclear. We herein investigated whether FxOH induces apoptosis in human pancreatic cancer cells. FxOH (5.0 µmol/L) significantly promoted the growth of human pancreatic cancer PANC-1 cells, but induced apoptosis in human colorectal cancer DLD-1 cells. A microarray-based gene analysis revealed that the gene sets of cell cycle, adhesion, PI3K/AKT, MAPK, NRF2, adipogenesis, TGF-ß, STAT, and Wnt signals in PANC-1 cells were markedly altered by FxOH. A western blot analysis showed that FxOH up-regulated the expression of integrin ß1 and PPARγ as well as the activation of pFAK(Tyr397), pPaxillin(Tyr31), and pAKT(Ser473) in PANC-1 cells, but exerted the opposite effects in DLD-1 cells. Moreover, the expression of FYN, a downstream target of integrin subunits, was up-regulated (7.4-fold by qPCR) in FxOH-treated PANC-1 cells. These results suggest that FxOH accelerates the growth of PANC-1 cells by up-regulating the expression of integrin ß1, FAK, Paxillin, FYN, AKT, and PPARγ.


Subject(s)
Pancreatic Neoplasms , Phosphatidylinositol 3-Kinases , Apoptosis , Carotenoids/pharmacology , Cell Line, Tumor , Humans , Pancreatic Neoplasms/drug therapy , beta Carotene/analogs & derivatives , beta Carotene/pharmacology
3.
Carcinogenesis ; 42(2): 210-219, 2021 02 25.
Article in English | MEDLINE | ID: mdl-32940665

ABSTRACT

Fucoxanthin (Fx), a marine carotenoid found in edible brown algae, is well known for having anticancer properties. The gut microbiota has been demonstrated as a hallmark for colorectal cancer progression in both humans and rodents. However, it remains unclear whether the gut microbiota is associated with the anticancer effect of Fx. We investigated the chemopreventive potency of Fx and its effect on gut microbiota in a mouse model of inflammation-associated colorectal cancer (by azoxymethane/dextran sulfate sodium treatment). Fx administration (30 mg/kg bw) during a 14 week period significantly inhibited the multiplicity of colorectal adenocarcinoma in mice. The number of apoptosis-like cleaved caspase-3high cells increased significantly in both colonic adenocarcinoma and mucosal crypts. Fx administration significantly suppressed Bacteroidlales (f_uc; g_uc) (0.3-fold) and Rikenellaceae (g_uc) (0.6-fold) and increased Lachnospiraceae (g_uc) (2.2-fold), compared with those of control mice. Oral administration of a fecal suspension obtained from Fx-treated mice, aimed to enhance Lachnospiraceae, suppress the number of colorectal adenocarcinomas in azoxymethane/dextran sulfate sodium-treated mice with a successful increase in Lachnospiraceae in the gut. Our findings suggested that an alteration in gut microbiota by dietary Fx might be an essential factor in the cancer chemopreventive effect of Fx in azoxymethane/dextran sulfate sodium-treated mice.


Subject(s)
Adenocarcinoma/prevention & control , Colitis, Ulcerative/drug therapy , Colitis-Associated Neoplasms/prevention & control , Gastrointestinal Microbiome/drug effects , Xanthophylls/administration & dosage , Adenocarcinoma/immunology , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Animals , Azoxymethane/administration & dosage , Azoxymethane/toxicity , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colitis-Associated Neoplasms/immunology , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Drug Screening Assays, Antitumor , Feces/microbiology , Gastrointestinal Microbiome/immunology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice
4.
Nutr Cancer ; 73(5): 889-898, 2021.
Article in English | MEDLINE | ID: mdl-33703973

ABSTRACT

Fucoxanthin is a marine xanthophyll found in edible brown algae, and a metabolite, fucoxanthinol (FxOH), possesses a potent apoptosis inducing effect in many cancer cells. Chloride intracellular channel 4 (CLIC4) is a member of the CLIC family that plays an important role in cancer development and apoptosis. However, the role of CLIC4 in FxOH-induced apoptosis is not well understood. In this study, we investigated whether CLIC4 affects the apoptotic properties of FxOH in human colorectal cancer (CRC) cells under FxOH treatment. Treating human CRC DLD-1 cells with 5.0 µmol/L FxOH significantly induced apoptosis. FxOH downregulated CLIC4, integrin ß1, NHERF2 and pSmad2 (Ser465/467) by 0.6-, 0.7-, 0.7-, and 0.5-fold, respectively, compared with control cells without alteration of Rab35 expression. No colocalizing change was observed in CLIC4-related proteins in either control or FxOH-treated cells. CLIC4 knockdown suppressed cell growth and apoptosis. Interestingly, apoptosis induction by FxOH almost disappeared with CLIC4 knockdown. Our findings suggested that CLIC4 could be involved in FxOH-induced apoptosis in human CRC.


Subject(s)
Colorectal Neoplasms , beta Carotene , Apoptosis , Cell Proliferation , Chloride Channels , Colorectal Neoplasms/drug therapy , Humans , beta Carotene/analogs & derivatives
5.
Int J Mol Sci ; 22(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34948416

ABSTRACT

Fucoxanthin (Fx) is a marine carotenoid with anti-inflammatory and anti-cancer properties in various animal models of carcinogenesis. However, there is currently no information on the effects of Fx in animal models of pancreatic cancer. We investigated the chemopreventive effects of Fx in C57BL/6J mice that received allogenic and orthotopic transplantations of cancer cells (KMPC44) derived from a pancreatic cancer murine model (Ptf1aCre/+; LSL-krasG12D/+). Using microarray, immunofluorescence, western blot, and siRNA analyses, alterations in cancer-related genes and protein expression were evaluated in pancreatic tumors of Fx-administered mice. Fx administration prevented the adenocarcinoma (ADC) development of pancreatic and parietal peritoneum tissues in a pancreatic cancer murine model, but not the incidence of ADC. Gene and protein expressions showed that the suppression of chemokine (C-C motif) ligand 21 (CCL21)/chemokine receptor 7 (CCR7) axis, its downstream of Rho A, B- and T-lymphocyte attenuator (BTLA), N-cadherin, αSMA, pFAK(Tyr397), and pPaxillin(Tyr31) were significantly suppressed in the pancreatic tumors of mice treated with Fx. In addition, Ccr7 knockdown significantly attenuated the growth of KMPC44 cells. These results suggest that Fx is a promising candidate for pancreatic cancer chemoprevention that mediates the suppression of the CCL21/CCR7 axis, BTLA, tumor microenvironment, epithelial mesenchymal transition, and adhesion.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinogenesis/drug effects , Pancreatic Neoplasms/prevention & control , Xanthophylls/therapeutic use , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Female , Mice, Inbred C57BL , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transcriptome/drug effects
6.
Biochem Biophys Res Commun ; 528(2): 305-310, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32475638

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is associated with hepatocyte injury, excessive oxidative stress, and chronic inflammation in fatty liver, and can progress to more severe liver diseases, such as cirrhosis and hepatocellular carcinoma. However, currently there are no effective therapies for NASH. Marine carotenoid, fucoxanthin (Fx), abundant in brown seaweeds, has variable biological properties, such as anti-cancer, anti-inflammatory, anti-oxidative and anti-obesity. However, the effect of Fx on the development of NASH has not been explored. We investigated the protective effects of Fx in diet-induced NASH model mice fed choline-deficient L-amino acid-defined high fat diet (CDAHFD). Fx administration significantly attenuated liver weight gain and hepatic fat accumulation, resulting in the alleviation of hepatic injury. Furthermore, the Fx-fed mice, not only exhibited reduced hepatic lipid oxidation, but also decreased mRNA expression levels of inflammation and infiltration-related genes compared to that of the CDAHFD-fed mice. Moreover, fucoxanthinol and amarouciaxanthin A, two Fx metabolites exerted anti-inflammatory effects in the liver via inhibiting the chemokine production in hepatocytes. In case of fibrosis, one of the features of advanced NASH, the expression of fibrogenic factors including activated-hepatic stellate cell marker was significantly decreased in the liver of Fx-fed mice. Thus, the present study elucidated that dietary Fx not only inhibited hepatic oxidative stress and inflammation but also prevented early phase of fibrosis in the diet-induced NASH model mice.


Subject(s)
Inflammation/pathology , Liver Cirrhosis/drug therapy , Liver/pathology , Non-alcoholic Fatty Liver Disease/drug therapy , Oxidative Stress , Xanthophylls/therapeutic use , Alanine Transaminase/blood , Amino Acids , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aspartate Aminotransferases/blood , Biomarkers/metabolism , Cell Line , Choline , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Inflammation/blood , Inflammation/complications , Inflammation/genetics , Lipid Metabolism/drug effects , Liver/drug effects , Liver/injuries , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/genetics , Male , Metabolome/drug effects , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/genetics , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xanthophylls/chemistry , Xanthophylls/pharmacology
7.
Arch Biochem Biophys ; 686: 108364, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32315653

ABSTRACT

Fucoxanthin (Fx), a major carotenoid found in brown seaweed, is known to show a unique and wide variety of biological activities. Upon absorption, Fx is metabolized to fucoxanthinol and amarouciaxanthin, and these metabolites mainly accumulate in visceral white adipose tissue (WAT). As seen in other carotenoids, Fx can quench singlet oxygen and scavenge a wide range of free radicals. The antioxidant activity is related to the neuroprotective, photoprotective, and hepatoprotective effects of Fx. Fx is also reported to show anti-cancer activity through the regulation of several biomolecules and signaling pathways that are involved in either cell cycle arrest, apoptosis, or metastasis suppression. Among the biological activities of Fx, anti-obesity is the most well-studied and most promising effect. This effect is primarily based on the upregulation of thermogenesis by uncoupling protein 1 expression and the increase in the metabolic rate induced by mitochondrial activation. In addition, Fx shows anti-diabetic effects by improving insulin resistance and promoting glucose utilization in skeletal muscle.


Subject(s)
Dietary Supplements/analysis , Seaweed/chemistry , Xanthophylls/chemistry , Xanthophylls/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/metabolism , Antioxidants/chemistry , Antioxidants/metabolism , Drug Discovery , Free Radicals/metabolism , Gene Expression Regulation/drug effects , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Insulin Resistance , Liver/metabolism , Molecular Structure , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Scavenger/metabolism , Singlet Oxygen/metabolism , Uncoupling Protein 1/chemistry , Uncoupling Protein 1/metabolism , Xanthophylls/adverse effects , beta Carotene/analogs & derivatives , beta Carotene/chemistry
8.
Mol Biol Rep ; 47(4): 2703-2711, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32180086

ABSTRACT

The combination of sarcopenia and obesity (i.e., sarcopenic obesity) is more strongly associated with disability and metabolic/cardiovascular diseases than obesity or sarcopenia alone. Therefore, countermeasures that simultaneously suppress fat gain and muscle atrophy to prevent an increase in sarcopenic obesity are warranted. The aim of this study was to investigate the simultaneous effects of fucoxanthinol (FXOH) on fat loss in mature adipocytes and the inhibition of atrophy and loss in myotubes induced by oxidative stress. C2C12 myotubes were treated with FXOH for 24 h and further incubated with hydrogen peroxide (H2O2) for 24 h. The area of myosin heavy chain-positive myotubes and the ROS concentration were measured. Mature 3T3-L1 adipocytes were treated with FXOH for 72 h. The triacylglycerol (TG) content and glycerol and fatty acid (FA) release were biochemically measured. The myotube area was smaller in H2O2-treated cells than that in control cells. However, FXOH protected against the H2O2-induced decreases in myotube area. Further, the ROS concentration was significantly higher in the FXOH-treated cells compared with that in the control cells, although it was significantly lower than that in the H2O2-treated cells. On the other hand, in the mature adipocytes, the TG content was significantly decreased by FXOH treatment compared to that in the control. Moreover, FXOH treatment significantly increased glycerol and FA release compared with that of the control. These results suggest that FXOH inhibits H2O2-induced atrophy and loss in myotubes and activates lipolysis and decreases the TG content in mature adipocytes. Accordingly, FXOH has the potential to exert anti-sarcopenic obesity effects.


Subject(s)
Muscular Atrophy/metabolism , Oxidative Stress/drug effects , beta Carotene/analogs & derivatives , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Atrophy/metabolism , Cell Line , Fatty Acids/metabolism , Hydrogen Peroxide/metabolism , Lipolysis/drug effects , Metabolic Diseases/pathology , Mice , Muscle Fibers, Skeletal/metabolism , Obesity/metabolism , Oxidative Stress/physiology , Triglycerides/metabolism , beta Carotene/metabolism , beta Carotene/pharmacology
9.
Mar Drugs ; 18(3)2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32143330

ABSTRACT

High-fat and high-sucrose diet (HFHSD)-induced obesity leads to oxidative stress and chronic inflammatory status. However, little is known about the beneficial effects of total lipids extracted from Spirulina. Hence, in the present study, Spirulina lipids were extracted with chloroform/methanol (SLC) or ethanol (SLE) and then their effects on oxidative stress and inflammation in the mice fed a HFHSD were investigated. The results show that the major lipid classes and fatty acid profiles of SLC and SLE were almost similar, but the gamma-linolenic acid (GLA) and carotenoid contents in SLE was a little higher than that in SLC. Dietary 4% SLC or SLE for 12 weeks effectively decreased the hepatic lipid hydroperoxide levels as well as increased the activities and mRNA levels of antioxidant enzymes in the mice fed a HFHSD. In addition, supplementation with SLC and SLE also markedly decreased the levels of serum pro-inflammatory cytokines and the mRNA expression of pro-inflammatory cytokines in the liver and epididymal white adipose tissue of mice fed a HFHSD, and the effects of SLC and SLE were comparable. These findings confirm for the first time that dietary Spirulina lipids could alleviate HFHSD-induced oxidative stress and inflammation.


Subject(s)
Inflammation/prevention & control , Lipids/pharmacology , Oxidative Stress/drug effects , Spirulina/chemistry , Animals , Antioxidants/pharmacology , Carotenoids/analysis , Carotenoids/pharmacology , Chloroform , Diet, High-Fat , Dietary Supplements , Ethanol , Fatty Acids/analysis , Fatty Acids/pharmacology , Lipids/chemistry , Liver/drug effects , Liver/metabolism , Male , Methanol , Mice , Mice, Inbred C57BL , Solvents , Sucrose , gamma-Linolenic Acid/analysis , gamma-Linolenic Acid/pharmacology
10.
Mar Drugs ; 17(11)2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31698797

ABSTRACT

The present study describes the variation in lipid components from 15 species of seaweeds belonging to the Chlorophyta, Ochrophyta, and Rhodophyta phyla collected in tropical (Indonesia) and temperate (Japan) areas. Analyses were performed of multiple components, including chlorophylls, carotenoids, n-3 and n-6 polyunsaturated fatty acids (PUFAs), and alpha tocopherol (α-Toc). Chlorophyll (Chl) and carotenoid contents varied among phyla, but not with the sampling location. Chl a and b were the major chlorophylls in Chlorophyta. Chl a and Chl c were the main chlorophylls in Ochrophyta, while Chl a was the dominant chlorophylls in Rhodophyta. ß-Carotene and fucoxanthin were detected as major seaweed carotenoids. The former was present in all species in a variety of ranges, while the latter was mainly found in Ochrophyta and in small quantities in Rhodophyta, but not in Chlorophyta. The total lipids (TL) content and fatty acids composition were strongly affected by sampling location. The TL and n-3 PUFAs levels tended to be higher in temperate seaweeds compared with those in tropical seaweeds. The major n-3 PUFAs in different phyla, namely, eicosapentaenoic acid (EPA) and stearidonic acid (SDA) in Ochrophyta, α-linolenic acid (ALA) and SDA in Chlorophyta, and EPA in Rhodophyta, accumulated in temperate seaweeds. Chlorophylls, their derivatives, and carotenoids are known to have health benefits, such as antioxidant activities, while n-3 PUFAs are known to be essential nutrients that positively influence human nutrition and health. Therefore, seaweed lipids could be used as a source of ingredients with health benefits for functional foods and nutraceuticals.


Subject(s)
Chlorophyta/chemistry , Lipids/chemistry , Rhodophyta/chemistry , Seaweed/chemistry , Carotenoids/chemistry , Carotenoids/isolation & purification , Chlorophyll/chemistry , Chlorophyll/isolation & purification , Dietary Supplements , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/isolation & purification , Indonesia , Japan , Lipids/isolation & purification
11.
J Clin Biochem Nutr ; 64(1): 52-58, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30705512

ABSTRACT

Fucoxanthin and its major metabolite, fucoxanthinol, have potent anti-cancer properties in carcinogenic model mice and against cancer cells. Evidence has accumulated regarding the diagnostic potential of biological metabolites as invasive and non-invasive obtainable approaches. We recently demonstrated that glycine was an effective predictor of the suppression of sphere formation and epithelial mesenchymal transition by fucoxanthinol in human colorectal cancer stem-like spheroids (colonospheres) under normoxia and hypoxia. In the present study, we investigated the suppressive effect of fucoxanthin on tumorigenesis derived from colonospheres in xenograft mice, and the alteration on the metabolite profiles of mouse tumors by fucoxanthin was evaluated. Fucoxanthin administration at 2.5 mg/kg body weight (p.o.) for 4 weeks significantly inhibited the incidence of tumors by inoculation of colonospheres suspension in BALB/c nu/nu mice compared with control mice, but not tumor sizes. In addition, fucoxanthin down-regulated tumor Cyclin D1 expression by 0.7-fold of that observed in the tumors of the control mice. Moreover, the tumor glycine level in the xenograft mice was decreased by fucoxanthin administration to 0.5-fold. These results imply the possibility of tumor metabolites as a prediction marker of tumorigenicity derived from colorectal cancer stem cells in mice.

12.
Mar Drugs ; 16(11)2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30388860

ABSTRACT

Carotenoids are natural pigments that contribute to light harvesting and photo-protection in photosynthetic organisms. In this study, we analyzed the carotenoid profiles, including mono-hydroxy and epoxy-carotenoids, in the economically valuable red seaweed Pyropia yezoensis, to clarify the detailed biosynthetic and metabolic pathways in the order Bangiales. P. yezoensis contained lutein, zeaxanthin, α-carotene, and ß-carotene, as major carotenoids in both the thallus and conchocelis stages. Monohydroxy intermediate carotenoids for the synthesis of lutein with an ε-ring from α-carotene, α-cryptoxanthin (ß,ε-caroten-3'-ol), and zeinoxanthin (ß,ε-caroten-3-ol) were identified. In addition, ß-cryptoxanthin, an intermediate in zeaxanthin synthesis from ß-carotene, was also detected. We also identified lutein-5,6-epoxide and antheraxanthin, which are metabolic products of epoxy conversion from lutein and zeaxanthin, respectively, by LC-MS and ¹H-NMR. This is the first report of monohydroxy-carotenoids with an ε-ring and 5,6-epoxy-carotenoids in Bangiales. These results provide new insights into the biosynthetic and metabolic pathways of carotenoids in red seaweeds.


Subject(s)
Biosynthetic Pathways , Carotenoids/analysis , Epoxy Compounds/metabolism , Seaweed/metabolism , Carotenoids/biosynthesis
13.
Nutr Cancer ; 69(7): 1043-1052, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28990814

ABSTRACT

Fucoxanthin (Fx), one of the major xanthophylls in brown algae, is known to be effective for colorectal cancer (CRC) chemoprevention through inhibiting cell growth, cell cycle and caspase activation. Recently, we observed fucoxanthinol (FuOH), an anti-cancer active metabolite of Fx, treatment of human CRC cells resulted in plenty of living floating cells several hours after exposure, and induced apoptosis. In the present study, we investigated whether FuOH induced anchorage-dependent apoptosis, that is "anoikis", along with integrin signal suppression in human CRC cells. We found that cells exposed to 2.5 µM FuOH clearly showed anti-proliferative and apoptotic effects to DLD-1 cells, human CRC cells. FuOH treatment of DLD-1 cells led to an increase in anoikis-like changes represented by Calcein AM negative/ethidium homodimer-1 positive cell and living floating cells. Moreover, FuOH decreased FAK activation, and altered integrin ß1 expression and distribution after 6 h treatment. After 24 h, the cells decreased PPARγ expression and Akt activation and increased integrin ß1 expression. Our findings suggested that FuOH can induce anoikis in CRC cells through suppression of integrin signals in human CRC cells.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , beta Carotene/analogs & derivatives , Allyl Compounds/pharmacology , Anoikis/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Curcumin/pharmacology , Dietary Supplements , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin beta1/metabolism , Isothiocyanates/pharmacology , Lipids/pharmacology , PPAR gamma , Sulfides/pharmacology , Sulfoxides , beta Carotene/pharmacology
14.
J Clin Biochem Nutr ; 61(1): 25-32, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28751806

ABSTRACT

Fucoxanthinol (FuOH), an intestinal metabolite form of fucoxanthin (Fx) isolated from marine algae, is known to possess multiple health benefits, such as prevention of human cancer. However, there is little available information about the effects of FuOH on colorectal cancer stem cells (CCSCs) and their contribution to drug resistance, tumorigenesis and cancer recurrence. In the present study, we investigated the anti-proliferative effect of FuOH on two putative CCSCs, CD44high/EpCAMhigh cells and colonospheres (Csps) formed by HT-29 human colorectal cancer cells, and the suppressive effects of FuOH on the growth of xenografted tumor. FuOH significantly inhibited the growth of CD44high/EpCAMhigh cells and disintegrated Csps and induced many condensed chromatin bodies in the cells in a dose-dependent manner. The IC50 value of FuOH for these changes in Csps was 1.8 µM. FuOH down-regulated pAkt (Ser473), PPARß/δ and PPARγ in Csps. These proteins play a critical role in cell proliferation, the cell cycle, metastasis and extracellular adhesion. Ten days after the administration of FuOH (5 mg/kg body weight) to the mice every 3 to 4 days significantly suppressed the Csps tumorigenesis when compared to the untreated control mice. Our results suggest that FuOH could be used as a chemopreventive agent against human CCSC.

15.
Mol Cell Biochem ; 417(1-2): 57-65, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27188184

ABSTRACT

Obesity and diabetes, risk factors for metabolic syndrome, are characterized by oxidative stress and inflammatory responses. Marine biofunctionals, astaxanthin (Ax) and squalene (SQ), were evaluated for their combined effect. Groups of male KK-A (y) mice were fed high fat/sucrose diet for 4 weeks, supplemented with either 0.1 %Ax, 2 %SQ or 0.1 %Ax + 2 %SQ. In comparison to control, Sod was elevated in only Ax + SQ. However, Gpx was highest in Ax + SQ, indicating the combined antioxidant effect of Ax and SQ. This was supported by elevated mRNA expression of Sod1 and Gpx1. Except adiponectin (elevated in Ax and Ax + SQ), expression of other inflammatory markers was not altered. Blood glucose levels were decreased in SQ and Ax + SQ while liver triglycerides decreased in SQ group. This is the first in vivo study demonstrating combined effects of Ax and SQ resulting in antioxidant effects and modulation of glucose/triglyceride levels. This study highlights the benefit of utilizing Ax and SQ together for management of obesity/diabetes.


Subject(s)
Oxidative Stress/drug effects , Squalene/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Liver/metabolism , Male , Mice , Mice, Obese , Obesity/blood , Obesity/drug therapy , Triglycerides/blood , Xanthophylls/pharmacology
16.
J Food Sci Technol ; 53(4): 2139-43, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27413244

ABSTRACT

Astaxanthin (Ax) and fucoxanthin/fucoxanthinol (FuOH) are marine xanthophylls exhibiting anti-oxidant effects. Squalene (SQ) is a triterpenoid and is a precursor of sterols. This study aimed to determine if SQ can improve the effect of Ax/FuOH on lipid peroxidation. RAW 264.7 cells were treated with different concentrations of Ax, FuOH and SQ and corresponding rate of cell survival was noted. In addition,combination groups - Ax + SQ and FuOH + SQ- were also run. Cells treated with Ax, FuOH, SQ, Ax + SQ and FuOH + SQ were stimulated with lipopolysaccharide and lipid hydroperoxides were estimated. Results showed that 5 µM Ax, 2 µM FuOH and 10 µM SQ supported cell survival. In presence of SQ, cell viability improved for higher concentrations of FuOH (5, 10 µM). Lipid hydroperoxides were supressed by Ax, FuOH, Ax + SQ and FUOH +SQ and were significantly lower in Ax + SQ, indicating the synergistic effect of Ax and SQ. To conclude, combination of Ax with SQ enhances its ability to supress lipid peroxidation while with FuOH, SQ attenuates the toxic effect at higher doses. Moreover, this is the first time that the combined effect of SQ and carotenoids has been studied and reported.

17.
Mar Drugs ; 13(8): 4799-813, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26248075

ABSTRACT

Fucoxanthin (Fx) is a marine carotenoid found in edible brown seaweeds. We previously reported that dietary Fx metabolite into fucoxanthinol (FxOH), attenuates the weight gain of white adipose tissue of diabetic/obese KK-Ay mice. In this study, to evaluate anti-diabetic effects of Fx, we investigated improving the effect of insulin resistance on the diabetic model of KK-Ay mice. Furthermore, preventing the effect of FxOH on low-grade chronic inflammation related to oxidative stress was evaluated on 3T3-L1 adipocyte cells and a RAW264.7 macrophage cell co-culture system. A diet containing 0.1% Fx was fed to diabetic model KK-Ay mice for three weeks, then glucose tolerance was observed. Fx diet significantly improved glucose tolerance compared with the control diet group.  In in vitro studies, FxOH showed suppressed tumor necrosis factor-α (TNF-α), and monocyte chemotactic protein-1 (MCP-1) mRNA expression and protein levels in a co-culture of adipocyte and macrophage cells. These findings suggest that Fx ameliorates glucose tolerance in the diabetic model mice. Furthermore, FxOH, a metabolite of Fx, suppresses low-grade chronic inflammation in adipocyte cells.


Subject(s)
Adipocytes/drug effects , Inflammation/drug therapy , Xanthophylls/pharmacology , beta Carotene/analogs & derivatives , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/physiopathology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Line , Chemokine CCL2/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diet , Disease Models, Animal , Inflammation/metabolism , Inflammation/physiopathology , Insulin Resistance/physiology , Macrophages/drug effects , Macrophages/metabolism , Mice , Obesity , RNA, Messenger/drug effects , Tumor Necrosis Factor-alpha/metabolism , Weight Gain/drug effects , beta Carotene/pharmacology
18.
J Agric Food Chem ; 72(8): 4142-4154, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38355398

ABSTRACT

The purpose of this study was to evaluate the preventive role and underlying mechanisms of fucoxanthin (Fx) on dextran sulfate sodium (DSS)-induced colitis in mice. The present data demonstrated that oral administration of Fx (50 and 200 mg/kg body weight/day) for 36 days significantly alleviated the severity of colitis in DSS-treated mice, as evidenced by attenuating body weight loss, bloody stool, diarrhea, shortened colon length, colonic epithelium distortion, a thin mucus layer, goblet cell depletion, damaged crypts, and extensive infiltration of inflammatory cells in the colonic mucosa. Additionally, Fx notably relieved DSS-induced intestinal epithelial barrier dysfunction via maintaining the tight junction function and preventing excessive apoptosis of colonic epithelial cells. Moreover, Fx effectively diminished colonic inflammation and oxidative stress in DSS-treated mice, and its mechanisms might be due to blunting the activation of NF-κB and NLRP3 inflammasome signaling pathways. Furthermore, Fx also modulates DSS-induced gut microbiota dysbiosis via recovering the richness and diversity of gut microbiota and reshaping the structure of gut microbiota, such as increasing the Firmicutes and Bacteroidota (F/B) ratio and elevating the relative abundance of some potential beneficial bacteria, including Lactobacillaceae and Lachnospiraceae. Overall, Fx might be developed as a promising functional ingredient to prevent colitis and maintain intestinal homeostasis.


Subject(s)
Colitis , Gastrointestinal Microbiome , Xanthophylls , Mice , Animals , Dextran Sulfate/adverse effects , Dysbiosis/chemically induced , Dysbiosis/drug therapy , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colon/metabolism , Mice, Inbred C57BL , Disease Models, Animal
19.
Food Funct ; 15(12): 6359-6373, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38787699

ABSTRACT

The aim of this study was to evaluate the preventive role and underlying mechanisms of fucoxanthin (Fx) on lipopolysaccharide (LPS)-induced intestinal barrier injury in mice. Our results demonstrated that the oral administration of Fx (50 and 200 mg per kg body weight per day) for consecutive 7 days significantly alleviated the severity of LPS-induced intestinal barrier injury in mice, as evidenced by attenuating body weight loss, improving intestinal permeability, and ameliorating intestinal morphological damage such as reduction in the ratio of the villus length to the crypt depth (V/C), intestinal epithelium distortion, goblet cell depletion, and low mucin 2 (MUC2) expression. Fx also significantly mitigated LPS-induced excessive apoptosis of intestinal epithelial cells (IECs) and curbed the decrease of tight junction proteins including claudin-1, occludin, and zonula occludens-1 in the ileum and colon. Additionally, Fx effectively alleviated LPS-induced extensive infiltration of macrophages and neutrophils into the intestinal mucosa, the overproduction of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 1beta (IL-1ß) and IL-6, and gasdermin D (GSDMD)-mediated pyroptosis of IECs. The underlying mechanisms might be associated with inhibiting the activation of nuclear factor-kappa B (NF-κB), mitogen-activated protein kinases (MAPKs) and nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathways. Moreover, Fx also notably restrained intestinal reactive oxygen species (ROS), malondialdehyde and protein carbonylation levels in LPS-treated mice, and it might be mediated by activating the nuclear factor-erythroid 2 related factor 2 (Nrf2) signaling pathway. Overall, these findings indicated that Fx might be developed as a potential effective dietary supplement to prevent intestinal barrier injury.


Subject(s)
Intestinal Mucosa , Lipopolysaccharides , Xanthophylls , Animals , Mice , Xanthophylls/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides/adverse effects , Male , Apoptosis/drug effects , NF-kappa B/metabolism , Permeability , Mice, Inbred C57BL , Tight Junction Proteins/metabolism , Cytokines/metabolism
20.
Biosci Biotechnol Biochem ; 77(3): 566-71, 2013.
Article in English | MEDLINE | ID: mdl-23470763

ABSTRACT

Lactoferrin (LF) is a multifunctional glycoprotein found in mammalian milk. We have shown in a previous clinical study that enteric-coated bovine LF tablets decreased visceral fat accumulation. To address the underlying mechanism, we conducted in vitro studies and revealed the anti-adipogenic action of LF in pre-adipocytes. The aim of this study was to assess whether LF could increase the lipolytic activity in mature adipocytes. Pre-adipocytes were prepared from rat mesenteric fat and differentiated into mature adipocytes for assays of lipolysis. The addition of LF significantly increased the glycerol concentration in the medium in a dose-dependent manner, whereas pepsin-degraded LF did not. A DNA microarray analysis demonstrated that LF decreased the expression of perilipin and affected the cAMP pathway. These findings are supported by the results of quantitative RT-PCR of perilipin and assays of cAMP. These data collectively indicate that visceral fat reduction by LF may result from the promotion of lipolysis and the additional anti-adipogenic activity of LF.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Cell Differentiation , Lactoferrin/pharmacology , Lipolysis/drug effects , Adipocytes/cytology , Animals , Cattle , Lactoferrin/metabolism , Lipolysis/genetics , Male , Oligonucleotide Array Sequence Analysis , Proteolysis , Rats , Rats, Sprague-Dawley , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL