Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nutr Res Pract ; 16(2): 161-172, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35392530

ABSTRACT

BACKGROUND/OBJECTIVES: Colorectal cancer (CRC) is the third most common cancer worldwide and has a high recurrence rate, which is associated with cancer stem cells (CSCs). ß-carotene (BC) possesses antioxidant activity and several anticancer mechanisms. However, no investigation has examined its effect on colon cancer stemness. MATERIALS/METHODS: CD133+CD44+ HCT116 and CD133+CD44+ HT-29 cells were isolated and analyzed their self-renewal capacity by clonogenic and sphere formation assays. Expressions of several CSCs markers and Wnt/ß-catenin signaling were examined. In addition, CD133+CD44+ HCT116 cells were subcutaneously injected in xenograft mice and analyzed the effect of BC on tumor formation, tumor volume, and CSCs markers in tumors. RESULTS: BC inhibited self-renewal capacity and CSC markers, including CD44, CD133, ALDH1A1, NOTCH1, Sox2, and ß-catenin in vitro. The effects of BC on CSC markers were confirmed in primary cells isolated from human CRC tumors. BC supplementation decreased the number and size of tumors and delayed the tumor-onset time in xenograft mice injected with CD133+CD44+ HCT116 cells. The inhibitory effect of BC on CSC markers and the Wnt/ß-catenin signaling pathway in tumors was confirmed in vivo as well. CONCLUSIONS: These results suggest that BC may be a potential therapeutic agent for colon cancer by targeting colon CSCs.

2.
Nutr Res Pract ; 14(5): 438-452, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33029285

ABSTRACT

BACKGROUND/OBJECTIVES: Brain senescence causes cognitive impairment and neurodegeneration. It has also been demonstrated that curcumin (Cur) and hesperetin (Hes), both antioxidant polyphenolic compounds, mediate anti-aging and neuroprotective effects. Therefore, the objective of this study was to investigate whether Cur, Hes, and/or their combination exert anti-aging effects in D-galactose (Dg)-induced aged neuronal cells and rats. MATERIALS/METHODS: SH-SY5Y cells differentiated in response to retinoic acid were treated with Cur (1 µM), Hes (1 µM), or a combination of both, followed by 300 mM Dg. Neuronal loss was subsequently evaluated by measuring average neurite length and analyzing expression of ß-tubulin III, phosphorylated extracellular signal-regulated kinases, and neurofilament heavy polypeptide. Cellular senescence and related proteins, p16 and p21, were also investigated, including their regulation of antioxidant enzymes. In vivo, brain aging was induced by injecting 250 mg/kg body weight (b.w.) Dg. The effects of supplementing this model with 50 mg/kg b.w. Cur, 50 mg/kg b.w. Hes, or a combination of both for 3 months were subsequently evaluated. Brain aging was examined with a step-through passive avoidance test and apoptosis markers were analyzed in brain cortex tissues. RESULTS: Cur, Hes, and their combination improved neuron length and cellular senescence by decreasing the number of ß-gal stained cells, down-regulated expression of p16 and p21, and up-regulated expression of antioxidant enzymes, including superoxide dismutase 1, glutathione peroxidase 1, and catalase. Administration of Cur, Hes, or their combination also tended to ameliorate cognitive impairment and suppress apoptosis in the cerebral cortex by down-regulating Bax and poly (ADP-ribose) polymerase expression and increasing Bcl-2 expression. CONCLUSIONS: Cur and Hes appear to attenuate Dg-induced brain aging via regulation of antioxidant enzymes and apoptosis. These results suggest that Cur and Hes may mediate neuroprotective effects in the aging process, and further study of these antioxidant polyphenolic compounds is warranted.

3.
J Nutr Biochem ; 82: 108402, 2020 08.
Article in English | MEDLINE | ID: mdl-32450500

ABSTRACT

The tumor microenvironment (TME), consisting of stromal fibroblasts, immune cells, cancer cells and other cell types, plays a crucial role in cancer progression and metastasis. M2 macrophages and activated fibroblasts (AFs) modulate behavior of cancer cells in the TME. Since nutritional effects on cancer progression, including colorectal cancer (CRC), may be mediated by alterations in the TME, we determined the ability of ß-carotene (BC) to mediate anti-cancer effects through regulation of macrophage polarization and fibroblast activation in CRC. The M2 macrophage phenotype was induced by treating U937 cells with phorbol-12-myristate-13-acetate and interleukin (IL)-4. Treatment of these M2 macrophages with BC led to suppression of M2-type macrophage-associated markers and of the IL-6/STAT3 signaling pathway. In separate experiments, AFs were induced by treating CCD-18Co cells with transforming growth factor-ß1. BC treatment suppressed expression of fibroblast activation markers. In addition, conditioned media from BC-treated M2 macrophages and AF inhibited cancer stem cell markers, colon cancer cell invasiveness and migration, and the epithelial-mesenchymal transition (EMT). In vivo, BC supplementation inhibited tumor formation and the expression of M2 macrophage markers in an azoxymethane/dextran sodium sulfate-induced colitis-associated CRC mouse model. To our knowledge, the present findings provide the first evidence suggesting that the potential therapeutic effects of BC on CRC are mediated by the inhibition of M2 macrophage polarization and fibroblast activation.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Fibroblasts/metabolism , Macrophages/metabolism , beta Carotene/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , HCT116 Cells , Humans , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , U937 Cells , beta Carotene/administration & dosage
4.
Eur J Nutr ; 58(4): 1603-1613, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29744610

ABSTRACT

PURPOSE: Walnuts (Juglans regia) are known to have anti-cancer and immunomodulatory effects. However, little information is available on the effects of walnut phenolic extract (WPE) on intestinal inflammation and colitis-associated colon cancer. METHODS: COLO205 cells were pretreated with WPE and then stimulated with tumor necrosis factor (TNF)-α. In the acute colitis model, wild type mice (C57BL/6) were administered 4% dextran sulfate sodium (DSS) for 5 days. In the chronic colitis model, interleukin (IL)-10-/- mice were administered with either the vehicle or WPE (20 mg/kg) by oral gavage daily for 2 weeks. In an inflammation-associated tumor model, wild type mice were administered a single intraperitoneal injection of azoxymethane followed by three cycles of 2% DSS for 5 days and 2 weeks of free water consumption. RESULTS: WPE significantly inhibited IL-8 and IL-1α expression in COLO205 cells. WPE attenuated both the TNF-α-induced IκB phosphorylation/degradation and NF-κB DNA binding activity. The administration of oral WPE significantly reduced the severity of colitis in both acute and chronic colitis models, including the IL-10-/- mice. In immunohistochemical staining, WPE attenuated NF-κB signaling in the colons of both colitis models. Finally, WPE also significantly reduced tumor development in a murine model of colitis-associated colon cancer (CAC). CONCLUSIONS: WPE ameliorates acute and chronic colitis and CAC in mice, suggesting that WPE may have potentials for the treatment of inflammatory bowel disease.


Subject(s)
Colonic Diseases/drug therapy , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Juglans , NF-kappa B/drug effects , Plant Extracts/pharmacology , Signal Transduction/drug effects , Animals , Colitis/drug therapy , Colitis/metabolism , Colonic Diseases/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Plant Extracts/administration & dosage
5.
Nutr Res Pract ; 12(2): 101-109, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29629026

ABSTRACT

BACKGROUND/OBJECTIVES: The objective of this study was to investigate the effects of vitamin C on inflammation, tumor development, and dysbiosis of intestinal microbiota in an azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced inflammation-associated early colon cancer mouse model. MATERIALS/METHODS: Male BALB/c mice were injected intraperitoneally with AOM [10 mg/kg body weight (b.w)] and given two 7-d cycles of 2% DSS drinking water with a 14 d inter-cycle interval. Vitamin C (60 mg/kg b.w. and 120 mg/kg b.w.) was supplemented by gavage for 5 weeks starting 2 d after the AOM injection. RESULTS: The vitamin C treatment suppressed inflammatory morbidity, as reflected by disease activity index (DAI) in recovery phase and inhibited shortening of the colon, and reduced histological damage. In addition, vitamin C supplementation suppressed mRNA levels of pro-inflammatory mediators and cytokines, including cyclooxygenase-2, microsomal prostaglandin E synthase-2, tumor necrosis factor-α, Interleukin (IL)-1ß, and IL-6, and reduced expression of the proliferation marker, proliferating cell nuclear antigen, compared to observations of AOM/DSS animals. Although the microbial composition did not differ significantly between the groups, administration of vitamin C improved the level of inflammation-related Lactococcus and JQ084893 to control levels. CONCLUSION: Vitamin C treatment provided moderate suppression of inflammation, proliferation, and certain inflammation-related dysbiosis in a murine model of colitis associated-early colon cancer. These findings support that vitamin C supplementation can benefit colonic health. Long-term clinical studies with various doses of vitamin C are warranted.

6.
Nutrients ; 8(12)2016 Dec 05.
Article in English | MEDLINE | ID: mdl-27929393

ABSTRACT

Type 2 diabetes is a major public health concern worldwide. Xylobiose (XB) consists of two molecules of d-xylose and is a major disaccharide in xylooligosaccharides that are used as prebiotics. We hypothesized that XB could regulate diabetes-related metabolic and genetic changes via microRNA expression in db/db mice. For six weeks, C57BL/KsJ-db/db mice received 5% XB as part of the total sucrose content of their diet. XB supplementation improved glucose tolerance with reduced levels of OGTT AUC, fasting blood glucose, HbA1c, insulin, and HOMA-IR. Furthermore, XB supplementation decreased the levels of total triglycerides, total cholesterol, and LDL-C. The expression levels of miR-122a and miR-33a were higher and lower in the XB group, respectively. In the liver, expressions of the lipogenic genes, including, fatty acid synthase (FAS), peroxisome proliferator activated receptor γ (PPARγ), sterol regulatory element-binding protein-1C (SREBP-1C), sterol regulatory element-binding protein-2 (SREBP-2), acetyl-CoA carboxylase (ACC), HMG-CoA reductase (HMGCR), ATP-binding cassette transporter G5/G8 (ABCG5/8), cholesterol 7 alpha-hydroxylase (CYP7A1), and sterol 12-alpha-hydroxylase (CYP8B1), as well as oxidative stress markers, including superoxide dismutase 1 (SOD1), superoxide dismutase 2 (SOD2), glutathione peroxidase (GPX), and catalase, were also regulated by XB supplementation. XB supplementation inhibited the mRNA expressions levels of the pro-inflammatory cytokines, tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, interleukin (IL)-6, and monocyte chemoattractant protein (MCP)-1, as well as phosphorylation of c-Jun N-terminal kinase/stress activated protein kinase (JNK/SAPK), p38 mitogen-activated protein kinases (MAPK), and extracellular signal-regulated kinases 1/2 (ERK1/2). These data demonstrate that XB exhibits anti-diabetic, hypolipogenic, and anti-inflammatory effects via regulation of the miR-122a/33a axis in db/db mice.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Disaccharides/administration & dosage , Lipogenesis/physiology , MicroRNAs/metabolism , Sweetening Agents/administration & dosage , Animals , Diabetes Mellitus, Experimental/metabolism , Dietary Supplements , Lipogenesis/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Prebiotics/administration & dosage
7.
Nutrients ; 8(7)2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27455311

ABSTRACT

Walnut has been known for its health benefits, including anti-cardiovascular disease and anti-oxidative properties. However, there is limited evidence elucidating its effects on cancer stem cells (CSCs) which represent a small subset of cancer cells that provide resistance against chemotherapy. This study aimed to evaluate the anti-CSCs potential of walnut phenolic extract (WPE) and its bioactive compounds, including (+)-catechin, chlorogenic acid, ellagic acid, and gallic acid. In the present study, CD133⁺CD44⁺ cells were isolated from HCT116 cells using fluorescence-activated cell sorting (FACS) and then treated with WPE. As a result, survival of the CD133⁺CD44⁺ HCT116 cells was inhibited and cell differentiation was induced by WPE. In addition, WPE down-regulated the CSC markers, CD133, CD44, DLK1, and Notch1, as well as the ß-catenin/p-GSK3ß signaling pathway. WPE suppressed the self-renewal capacity of CSCs. Furthermore, the WPE exhibited stronger anti-CSC effects than its individual bioactive compounds. Finally, the WPE inhibited specific CSC markers in primary colon cancer cells isolated from primary colon tumor. These results suggest that WPE can suppress colon cancer by regulating the characteristics of colon CSCs.


Subject(s)
Anticarcinogenic Agents/metabolism , Antineoplastic Agents, Phytogenic/metabolism , Colorectal Neoplasms/prevention & control , Juglans/chemistry , Neoplastic Stem Cells/metabolism , Nuts/chemistry , Plant Extracts/metabolism , Anticarcinogenic Agents/analysis , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/isolation & purification , Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Biomarkers, Tumor/metabolism , Catechin/analysis , Catechin/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Chlorogenic Acid/analysis , Chlorogenic Acid/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dietary Supplements , Ellagic Acid/analysis , Ellagic Acid/metabolism , Gallic Acid/analysis , Gallic Acid/metabolism , HCT116 Cells , Humans , Neoplastic Stem Cells/pathology , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Republic of Korea , Tumor Cells, Cultured
8.
Nutr Res ; 34(10): 894-905, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25287291

ABSTRACT

Sasa quelpaertensis leaves exert anti-inflammatory and anticarcinogenic effects, although it remains unclear whether these leaves can suppress inflammation-related intestinal diseases. This study hypothesized that Sasa quelpaertensis leaf extract (SQE) exerts a protective effect against inflammation in a dextran sulfate sodium (DSS)-induced colitis mouse model. Therefore, colon tissues of DSS-induced colitis mice that were treated with SQE were assayed for levels of proinflammatory markers, mitogen-activated protein kinase signaling, and activation of nuclear factor κB. For this purpose, mice were pretreated with SQE (100 mg/kg or 300 mg/kg body weight) by gavage for a 2-week period. Mice then received either SQE or sulfasalazine (100 mg/kg body weight) with 2.5% DSS in drinking water for 7 days twice daily and 7 days of tap water ad libitum between DSS treatment. Treatment with SQE was found to attenuate the severity of DSS-induced colitis, as assessed by disease activity index scores, shrinkage of colon length, and histopathologic changes. SQE reduced DSS-induced proliferation in distal colon tissues. It also significantly suppressed levels of tumor necrosis factor-α in serum and colon tissues, nitric oxide synthase, cyclooxygenase, and levels of phosphorylated c-Jun N-terminal kinases, p38, extracellular-signal-regulated kinases 1/2, and IκBα in colon tissues. To our knowledge, this is the first study to demonstrate that SQE supplementation can exert an anti-inflammatory effect on experimental chronic colitis.


Subject(s)
Colitis/drug therapy , Colon/drug effects , Inflammation Mediators/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Sasa , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cyclooxygenase 2/metabolism , Dextran Sulfate , I-kappa B Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-KappaB Inhibitor alpha , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Plant Extracts/pharmacology , Plant Leaves , Severity of Illness Index , Tumor Necrosis Factor-alpha/metabolism
9.
Biochem Biophys Res Commun ; 450(4): 1475-80, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25019987

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid cancer in young children and malignant NB cells have been shown to possess cancer stem cell (CSC) characteristics. Thus, the successful elimination of CSCs represents a strategy for developing an effective preventive and chemotherapeutic agent. CSCs are characterized by differentiation and tumorigenicity. ß-Carotene (BC) has been associated with many anticancer mechanisms, although the efficacy of BC on CSCs remains unclear. In the present study, the effects of BC on tumor cell differentiation and tumorigenicity was investigated using a xenograft model. Mice were pretreated with BC for 21 days, then received a subcutaneous injection of SK-N-BE(2)C cells. Both tumor incidence and tumor growth were significantly inhibited for mice that received BC supplementation compared to the control group. Treatment with BC has also been shown to induce tumor cell differentiation by up-regulating differentiation markers, such as vimentin, peripherin, and neurofilament. Conversely, BC treatment has been shown to significantly suppress tumor stemness by down-regulating CSC markers such as Oct 3/4 and DLK1. BC treatment also significantly down-regulated HIF1-α expression and its downstream target, vascular endothelial growth factor (VEGF). Taken together, these results suggest that BC is a potential chemotherapeutic reagent for the treatment of NB, and mediates this effect by regulating the differentiation and stemness of CSCs, respectively.


Subject(s)
Cell Differentiation/drug effects , Neoplastic Stem Cells/drug effects , Neuroblastoma/pathology , beta Carotene/pharmacology , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neuroblastoma/metabolism , Polymerase Chain Reaction , beta Carotene/administration & dosage
10.
Integr Cancer Ther ; 13(6): 529-40, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24838270

ABSTRACT

Lung cancer is the leading cause of cancer death worldwide, and most chemotherapeutic drugs have limited success in treating this disease. Furthermore, some drugs show undesirable side effects due to the enrichment of cancer stem cells (CSCs) that are present, leading to resistance to conventional chemotherapy and tumor relapse. CSCs possess self-renewal characteristics, aggressive tumor initiating activity, and ability to facilitate tumor metastasis. Therefore, development of nontoxic agents that can potentiate chemotherapy and eliminate CSCs would be highly desirable. In the present study, we investigated whether Sasa quelpaertensis leaf extracts (SQE) and cisplatin (CIS), individually or in combination, would exert anti-CSC and antimetastatic effect in H1299 and A549 human lung cancer cells. Following these treatments, cell growth, phosphorylation of phosphoinositide-3 kinase, and activation of the mammalian target of rapamycin were inhibited. Decreased serial sphere formation, clonogenicity, and expression of major stem cell markers, such as CD44 and SOX-2, in CD44(+) cancer stem cells were also observed. In addition, inhibition of cell migration and invasion in both cell lines as well as inhibition of matrix metalloproteinase-2 activity and expression were detected. Importantly, the anticancer stemness and antimetastasis effects in each of these assays were greater for the combined treatment with SQE and CIS than with each treatment individually. In conclusion, the data suggest that SQE alone, or in combination with CIS, represents a promising therapeutic strategy for eliminating cancer stemness and cell invasion potential of CSCs, thereby treating and preventing metastatic lung cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology , Sasa/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/administration & dosage , Drug Synergism , Humans , Lung Neoplasms/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Plant Extracts/administration & dosage , Plant Leaves
11.
J Med Food ; 17(5): 571-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24738745

ABSTRACT

Sasa quelpaertensis is a bamboo leaf that is only grown on Jeju Island in South Korea. It is used as a bamboo tea that is consumed for therapeutic purposes, particularly for its anti-diabetic, diuretic, and anti-inflammatory effects. This study investigated the effect of S. quelpaertensis leaf extract (SQE) on high fat-induced lipid abnormalities and regulation of lipid metabolism-related gene expressions in rats. SQE supplementation significantly decreased the levels of plasma triglycerides, total cholesterol, and low-density lipoprotein cholesterol as well as the atherogenic index. SQE restored levels of plasma high-density lipoprotein cholesterol, which were lowered by a high fat diet. Plasma and cardiac resistin levels were also significantly decreased by SQE supplementation. In adipose tissue, mRNA levels of CAAT/enhancer-binding protein ß (C/EBPß) were suppressed in the SQE group. SQE supplementation decreased the accumulation of lipid droplets, inflammatory cell infiltrations, levels of triglycerides, and total lipids in the liver and effectively down-regulated expression of sterol regulatory element binding protein-1 (SREBP-1), fatty acid synthetase (FAS), and uncoupling protein 2 (UCP-2). These results suggest that SQE may be a potential treatment for high fat-related disorders by improving lipid profiles and modulating lipid metabolism.


Subject(s)
Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Hyperlipidemias/drug therapy , Lipid Metabolism/genetics , Plant Extracts/therapeutic use , Sasa/chemistry , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents , Cholesterol/blood , Diet , Down-Regulation , Fatty Acid Synthases/genetics , Hyperlipidemias/blood , Ion Channels/genetics , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Liver/metabolism , Male , Mitochondrial Proteins/genetics , Phytotherapy , Plant Leaves/chemistry , Rats , Rats, Sprague-Dawley , Republic of Korea , Resistin/analysis , Sterol Regulatory Element Binding Protein 1/genetics , Triglycerides/blood , Uncoupling Protein 2
12.
J Nutr Biochem ; 25(6): 655-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24746828

ABSTRACT

Neuroblastoma is the most prevalent extracranial solid tumor in childhood and has poor clinical outcome due to its high potential for metastasis. Consequently, an understanding of the mechanisms that modulate cancer cell invasion, migration and metastasis is important for the development of more effective chemotherapeutic agents. While ß-carotene is a vitamin A precursor that has been shown to exert antioxidant and anticancer effects, the anti-metastatic effects of ß-carotene on neuroblastoma cells remain poorly understood. The aim of the present study was to investigate the anti-metastatic effects of ß-carotene on highly malignant SK-N-BE(2)C neuroblastoma cells in vitro and in vivo. Treatment of SK-N-BE(2)C cells with ß-carotene was found to attenuate the migratory and invasive capabilities of the cells. In addition, the enzymatic activity and expression of matrix metalloproteinase (MMP)-2 was suppressed following ß-carotene treatment under both normoxia and hypoxia. To induce metastasis, immunodeficient nude mice were injected with SK-N-BE(2)C cells via the tail vein in vivo. The incidence of liver metastasis and mean tumor volume in mice that were administered ß-carotene was decreased compared to controls. Furthermore, mRNA levels of MMPs, membrane-type (MT) 2 MMP and tissue inhibitors of metalloproteinases in liver tumor tissues were also lower following ß-carotene treatment. Level of hypoxia-inducible factor-1α (HIF-1α) and its downstream targets, vascular endothelial growth factor and glucose transporter 1 (GLUT1), were lower both in vitro and in vivo following ß-carotene treatment. In conclusion, the present study provides the first evidence that ß-carotene may represent an effective chemotherapeutic agent by regulating the invasion and metastasis of neuroblastoma via HIF-1α.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Dietary Supplements , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neuroblastoma/prevention & control , beta Carotene/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Male , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroblastoma/secondary , Random Allocation , Specific Pathogen-Free Organisms , Tissue Inhibitor of Metalloproteinases/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Tumor Burden , Xenograft Model Antitumor Assays , beta Carotene/metabolism
13.
Biosci Biotechnol Biochem ; 77(4): 722-8, 2013.
Article in English | MEDLINE | ID: mdl-23563563

ABSTRACT

A neuroblastoma is an extracranial solid tumor diagnosed in childhood. Since tumor metastasis is the main cause of death for most neuroblastoma patients, an understanding of the mechanisms that modulate cancer cell invasion is a key to developing more effective chemotherapeutic agents. In the current study, we examined to determine whether mulberry leaf (ML) extract effectively inhibits the invasion potential of neuroblastoma cells in vitro. ML extract was found to suppress cell invasiveness as well as the activity and expression of matrix metalloproteinase-2 (MMP-2) under both normoxia and hypoxia in neuroblastoma. ML extract downregulated the expression of hypoxia-inducible factor-1α (HIF-1α), a well-known regulator of tumor metastasis, and its downstream targets, vascular endothelial growth factor (VEGF) and glucose transporter-1 (GLUT-1). Taken together, these results suggest that ML extract has chemotherapeutic effects on neuroblastoma cells by regulating invasion potential, thereby controlling the metastasis of neuroblastomas.


Subject(s)
Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Morus/chemistry , Neuroblastoma/pathology , Plant Extracts/pharmacology , Plant Leaves/chemistry , Antineoplastic Agents/isolation & purification , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Plant Extracts/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL