Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Steroid Biochem Mol Biol ; 231: 106307, 2023 07.
Article in English | MEDLINE | ID: mdl-37030416

ABSTRACT

K vitamins are well known as essential cofactors for hepatic γ-carboxylation of coagulation factors, but their potential role in chronic diseases including cancer is understudied. K2, the most abundant form of vitamin K in tissues, exerts anti-cancer effects via diverse mechanisms which are not completely understood. Our studies were prompted by previous work demonstrating that the K2 precursor menadione synergized with 1,25 dihydroxyvitamin D3 (1,25(OH)2D3) to inhibit growth of MCF7 luminal breast cancer cells. Here we assessed whether K2 modified the anti-cancer effects of 1,25(OH)2D3 in triple negative breast cancer (TNBC) cell models. We examined the independent and combined effects of these vitamins on morphology, cell viability, mammosphere formation, cell cycle, apoptosis and protein expression in three TNBC cell models (MDA-MB-453, SUM159PT, Hs578T). We found that all three TNBC cell lines expressed low levels of the vitamin D receptor (VDR) and were modestly growth inhibited by 1,25(OH)2D3 in association with cell cycle arrest in G0/G1. Induction of differentiated morphology by 1,25(OH)2D3 was observed in two of the cell lines (MDA-MB-453, Hs578T). Treatment with K2 alone reduced viability of MDA-MB-453 and SUM159PT cells but not Hs578T cells. Co-treatment with 1,25(OH)2D3 and K2 significantly reduced viable cell number relative to either treatment alone in Hs578T and SUM159PT cells. The combination treatment induced G0/G1 arrest in MDA-MB-453 cells, Hs578T and SUM159PT cells. Combination treatment altered mammosphere size and morphology in a cell specific manner. Of particular interest, treatment with K2 increased VDR expression in SUM159PT cells suggesting that the synergistic effects in these cells may be secondary to increased sensitivity to 1,25(OH)2D3. The phenotypic effects of K2 in TNBC cells did not correlate with γ-carboxylation suggesting non-canonical actions. In summary, 1,25(OH)2D3 and K2 exert tumor suppressive effects in TNBC cells, inducing cell cycle arrest leading to differentiation and/or apoptosis depending on the specific cell line. Further mechanistic studies to clarify common and unique targets of these two fat soluble vitamins in TNBC are warranted.


Subject(s)
Calcitriol , Triple Negative Breast Neoplasms , Humans , Calcitriol/pharmacology , Vitamin K 2/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Cells, Cultured , Receptors, Calcitriol/metabolism , Vitamin K , Vitamins/pharmacology
2.
Trends Mol Med ; 28(10): 864-881, 2022 10.
Article in English | MEDLINE | ID: mdl-36028390

ABSTRACT

Phylloquinone (vitamin K1) and menaquinones (vitamin K2 family) are essential for post-translational γ-carboxylation of a small number of proteins, including clotting factors. These modified proteins have now been implicated in diverse physiological and pathological processes including cancer. Vitamin K intake has been inversely associated with cancer incidence and mortality in observational studies. Newly discovered functions of vitamin K in cancer cells include activation of the steroid and xenobiotic receptor (SXR) and regulation of oxidative stress, apoptosis, and autophagy. We provide an update of vitamin K biology, non-canonical mechanisms of vitamin K actions, the potential functions of vitamin K-dependent proteins in cancer, and observational trials on vitamin K intake and cancer.


Subject(s)
Neoplasms , Vitamin K , Biology , Humans , Neoplasms/etiology , Pregnane X Receptor , Proteins , Vitamin K/metabolism , Vitamin K 1/metabolism , Vitamin K 2/metabolism
3.
Cancer Prev Res (Phila) ; 13(8): 673-686, 2020 08.
Article in English | MEDLINE | ID: mdl-32467291

ABSTRACT

Ductal carcinoma in situ (DCIS), which accounts for one out of every five new breast cancer diagnoses, will progress to potentially lethal invasive ductal carcinoma (IDC) in about 50% of cases. Vitamin D compounds have been shown to inhibit progression to IDC in the MCF10DCIS model. This inhibition appears to involve a reduction in the cancer stem cell-like population in MCF10DCIS tumors. To identify genes that are involved in the vitamin D effects, a global transcriptomic analysis was undertaken of MCF10DCIS cells grown in mammosphere cultures, in which cancer stem-like cells grow preferentially and produce colonies by self-renewal and maturation, in the presence and absence of 1α25(OH)2D3 and a vitamin D analog, BXL0124. Using next-generation RNA-sequencing, we found that vitamin D compounds downregulated genes involved in maintenance of breast cancer stem-like cells (e.g., GDF15), epithelial-mesenchymal transition, invasion, and metastasis (e.g., LCN2 and S100A4), and chemoresistance (e.g., NGFR, PPP1R1B, and AGR2), while upregulating genes associated with a basal-like phenotype (e.g., KRT6A and KRT5) and negative regulators of breast tumorigenesis (e.g., EMP1). Gene methylation status was analyzed to determine whether the changes in expression induced by vitamin D compounds occurred via this mechanism. Ingenuity pathway analysis was performed to identify upstream regulators and downstream signaling pathway genes differentially regulated by vitamin D, including TP63 and vitamin D receptor -mediated canonical pathways in particular. This study provides a global profiling of changes in the gene signature of DCIS regulated by vitamin D compounds and possible targets for chemoprevention of DCIS progression to IDC in patients.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/prevention & control , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Neoplastic Stem Cells/drug effects , Vitamin D/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation/drug effects , Datasets as Topic , Disease Progression , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplastic Stem Cells/pathology , RNA-Seq , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Vitamin D/analogs & derivatives
4.
J Med Food ; 22(6): 578-586, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30864851

ABSTRACT

Our previous studies have demonstrated antioxidant and cytoprotective properties of red ginseng oil (RGO). However, the role of RGO in models of intestinal inflammation has not been elucidated. In this study, we evaluated the chemopreventive effect of RGO in a mouse model of azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colitis and explored its underlying mechanisms. Male C57BL/6 mice were intraperitoneally injected with a single dose of AOM (10 mg/kg), followed by 1.5% DSS in drinking water for 7 days to produce colon carcinogenesis. RGO at 10 or 100 mg/kg was orally given for 17 weeks. RGO supplementation reduced the plasma nitric oxide (NO) concentration as well as lipid peroxidation and inhibited the production of proinflammatory factors such as inducible NO synthase, cyclooxygenase-2, interleukin 1ß, IL-6, and tumor necrosis factor-α in the mouse colitis tissue. Increased phosphorylation levels of p65 and IκB by AOM/DSS exposure were attenuated by the presence of RGO. In addition, RGO supplementation induced the activity of primary antioxidant enzymes such as superoxide dismutase and catalase as well as the expression of nuclear factor erythroid 2-related factor 2-mediated antioxidant enzyme hemeoxygenase-1 in the colons of AOM/DSS-treated mice. These findings indicate that RGO may be a potent natural chemopreventive agent for ameliorating inflammatory bowel diseases.


Subject(s)
Colitis/drug therapy , Colonic Neoplasms/prevention & control , Panax/chemistry , Plant Extracts/administration & dosage , Animals , Azoxymethane/adverse effects , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colon/drug effects , Colon/immunology , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , Plant Extracts/isolation & purification , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
Carcinogenesis ; 39(8): 1045-1055, 2018 07 30.
Article in English | MEDLINE | ID: mdl-29846560

ABSTRACT

Estrogen plays an important role in breast cancer development. While the mechanism of the estrogen effects is not fully elucidated, one possible route is by increasing the stem cell-like properties in the tumors. Tocopherols are known to reduce breast cancer development and progression. The aim of the present study is to investigate the effects of tocopherols on the regulation of breast cancer stemness mediated by estrogen. To determine the effects of tocopherols on estrogen-influenced breast cancer stem cells, the MCF-7 tumorsphere culture system, which enriches for mammary progenitor cells and putative breast cancer stem cells, was utilized. Treatment with estrogen resulted in an increase in the CD44+/CD24- subpopulation and aldehyde dehydrogenase activity in tumorspheres as well as the number and size of tumorspheres. Tocopherols inhibited the estrogen-induced expansion of the breast cancer stem population. Tocopherols decreased the levels of stem cell markers, including octamer-binding transcription factor 4 (OCT4), CD44 and SOX-2, as well as estrogen-related markers, such as trefoil factor (TFF)/pS2, cathepsin D, progesterone receptor and SERPINA1, in estrogen-stimulated tumorspheres. Overexpression of OCT4 increased CD44 and sex-determining region Y-box-2 levels and significantly increased cell invasion and expression of the invasion markers, matrix metalloproteinases, tissue inhibitors of metalloproteinase and urokinase plasminogen activator, and tocopherols inhibited these OCT4-mediated effects. These results suggest a potential inhibitory mechanism of tocopherols in estrogen-induced stemness and cell invasion in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Estrogens/metabolism , Neoplastic Stem Cells/drug effects , Octamer Transcription Factor-3/metabolism , Tocopherols/pharmacology , Breast Neoplasms/pathology , Cell Movement/drug effects , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplastic Stem Cells/metabolism , Receptors, Estrogen/metabolism , Tocopherols/therapeutic use
6.
Cancer Prev Res (Phila) ; 10(12): 694-703, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28972008

ABSTRACT

Despite experimental evidence elucidating the antitumor activities of tocopherols, clinical trials with α-tocopherol (α-T) have failed to demonstrate its beneficial effects in cancer prevention. This study compared the chemopreventive efficacy of individual tocopherols (α-, δ-, and γ-T) and a γ-T-rich tocopherol mixture (γ-TmT) in the August-Copenhagen Irish (ACI) rat model of estrogen-mediated mammary cancer. Female ACI rats receiving 17ß-estradiol (E2) implants were administered with 0.2% α-T, δ-T, γ-T, or γ-TmT for 30 weeks. Although α-T had no significant effects on mammary tumor growth in ACI rats, δ-T, γ-T, and γ-TmT reduced mammary tumor volume by 51% (P < 0.05), 60% (P < 0.01), and 59% (P < 0.01), respectively. Immunohistochemical analysis revealed that δ-T, γ-T, and γ-TmT reduced levels of the cell proliferation marker, proliferating cell nuclear antigen, in the rat mammary tumors. To gain further insight into the biological functions of different forms of tocopherols, RNA-seq analysis of the tumors was performed. Treatment with γ-T induced robust gene expression changes in the mammary tumors of ACI rats. Ingenuity Pathway Analysis identified "Cancer" as a top disease pathway and "Tumor growth" and "Metastasis" as the top signaling pathways modulated by γ-T. Although the results need further functional validation, this study presents an unbiased attempt to understand the differences between biological activities of individual forms of tocopherols at the whole transcriptome level. In conclusion, δ-T and γ-T have superior cancer preventive properties compared to α-T in the prevention of estrogen-mediated mammary carcinogenesis. Cancer Prev Res; 10(12); 694-703. ©2017 AACR.


Subject(s)
Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/drug therapy , Tocopherols/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Apoptosis , Carcinogenesis , Cell Adhesion , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Disease Progression , Extracellular Matrix/metabolism , Female , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Metastasis , Neovascularization, Pathologic , Rats , Rats, Inbred ACI , Signal Transduction , alpha-Tocopherol/pharmacology , gamma-Tocopherol/pharmacology
7.
Molecules ; 22(9)2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28885585

ABSTRACT

Hair loss (alopecia) is a universal problem for numerous people in the world. The present study was conducted to investigate the effects of red ginseng oil (RGO) and its major components on hair re-growth using testosterone (TES)-induced delay of anagen entry in C57BL/6 mice and their mechanisms of action. Seven-week-old C57BL/6 mice were daily treated with TES for 1 h prior to topical application of 10% RGO, 1% linoleic acid (LA), 1% ß-sitosterol (SITOS), or 1% bicyclo(10.1.0)tridec-1-ene (BICYCLO) once a day for 28 days. Hair regenerative capacity was significantly restored by treatment of RGO and its major compounds in the TES-treated mice. Histological analysis showed that RGO along with LA and SITOS but not BICYCLO promoted hair growth through early inducing anagen phase that was delayed by TES in mice. Treatment of mice with RGO, LA, or SITOS up-regulated Wnt/ß-catenin and Shh/Gli pathways-mediated expression of genes such as ß-catenin, Lef-1, Sonic hedgehog, Smoothened, Gli-1, Cyclin D1, and Cyclin E in the TES-treated mice. In addition, RGO and its major components reduced the protein level of TGF-ß but enhanced the expression of anti-apoptotic protein Bcl-2. These results suggest that RGO is a potent novel therapeutic natural product for treatment of androgenic alopecia possibly through hair re-growth activity of its major components such as LA and SITOS.


Subject(s)
Alopecia/drug therapy , Hair Follicle/drug effects , Linoleic Acid/pharmacology , Panax/chemistry , Plant Oils/pharmacology , Sitosterols/pharmacology , Alopecia/chemically induced , Alopecia/genetics , Alopecia/pathology , Animals , Cyclins/genetics , Cyclins/metabolism , Disease Models, Animal , Gene Expression Regulation , Hair Follicle/growth & development , Hair Follicle/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Mice, Inbred C57BL , Plant Oils/isolation & purification , Proto-Oncogene Proteins c-bcl-2/agonists , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Regeneration/drug effects , Regeneration/genetics , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Testosterone/administration & dosage , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , beta Catenin/genetics , beta Catenin/metabolism
8.
Cancer Prev Res (Phila) ; 10(3): 188-197, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28096236

ABSTRACT

Estrogens have been implicated as complete carcinogens for breast and other tissues through mechanisms involving increased cell proliferation, oxidative stress, and DNA damage. Because of their potent antioxidant activity and other effects, tocopherols have been shown to exert antitumor activities in various cancers. However, limited information is available on the effect of different forms of tocopherols in estrogen-mediated breast cancer. To address this, we examined the effects of α-, γ-, and δ-tocopherols as well as a natural γ-tocopherol-rich mixture of tocopherols, γ-TmT, on estrogen-stimulated MCF-7 cells in vitro and in vivo For the in vivo studies, MCF-7 cells were injected into the mammary fat pad of immunodeficient mice previously implanted with estrogen pellets. Mice were then administered diets containing 0.2% α-, γ-, δ-tocopherol, or γ-TmT for 5 weeks. Treatment with α-, γ-, δ-tocopherols, and γ-TmT reduced tumor volumes by 29% (P < 0.05), 45% (P < 0.05), 41% (P < 0.05), and 58% (P < 0.01), as well as tumor weights by 20%, 37% (P < 0.05), 39% (P < 0.05), and 52% (P < 0.05), respectively. γ- and δ-tocopherols and γ-TmT inhibited the expression of cell proliferation-related genes such as cyclin D1 and c-Myc, and estrogen-related genes such as TFF/pS2, cathepsin D, and progesterone receptor in estrogen-stimulated MCF-7 cells in vitro Further, γ- and δ-tocopherols decreased the levels of estrogen-induced oxidative stress and nitrosative stress markers, 8-hydroxy-2'-deoxyguanosine and nitrotyrosine, as well as the DNA damage marker, γ-H2AX. Our results suggest that γ- and δ-tocopherols and the γ-tocopherol-rich mixture are effective natural agents for the prevention and treatment of estrogen-mediated breast cancer. Cancer Prev Res; 10(3); 188-97. ©2017 AACR.


Subject(s)
Antioxidants/pharmacology , Breast Neoplasms/pathology , Oxidative Stress/drug effects , Tocopherols/pharmacology , gamma-Tocopherol/pharmacology , Animals , Cell Proliferation/drug effects , Estrogens/toxicity , Female , Humans , MCF-7 Cells , Mice, Nude , Xenograft Model Antitumor Assays
9.
J Steroid Biochem Mol Biol ; 173: 122-129, 2017 10.
Article in English | MEDLINE | ID: mdl-27923595

ABSTRACT

Triple-negative breast cancer is one of the least responsive breast cancer subtypes to available targeted therapies due to the absence of hormonal receptors, aggressive phenotypes, and the high rate of relapse. Early breast cancer prevention may therefore play an important role in delaying the progression of triple-negative breast cancer. Cancer stem cells are a subset of cancer cells that are thought to be responsible for tumor progression, treatment resistance, and metastasis. We have previously shown that vitamin D compounds, including a Gemini vitamin D analog BXL0124, suppress progression of ductal carcinoma in situ in vivo and inhibit cancer stem-like cells in MCF10DCIS mammosphere cultures. In the present study, the effects of vitamin D compounds in regulating breast cancer stem-like cells and differentiation in triple-negative breast cancer were assessed. Mammosphere cultures, which enriches for breast cancer cells with stem-like properties, were used to assess the effects of 1α,25(OH)2D3 and BXL0124 on cancer stem cell markers in the triple-negative breast cancer cell line, SUM159. Vitamin D compounds significantly reduced the mammosphere forming efficiency in primary, secondary and tertiary passages of mammospheres compared to control groups. Key markers of cancer stem-like phenotype and pluripotency were analyzed in mammospheres treated with 1α,25(OH)2D3 and BXL0124. As a result, OCT4, CD44 and LAMA5 levels were decreased. The vitamin D compounds also down-regulated the Notch signaling molecules, Notch1, Notch2, Notch3, JAG1, JAG2, HES1 and NFκB, which are involved in breast cancer stem cell maintenance. In addition, the vitamin D compounds up-regulated myoepithelial differentiating markers, cytokeratin 14 and smooth muscle actin, and down-regulated the luminal marker, cytokeratin 18. Cytokeratin 5, a biomarker associated with basal-like breast cancer, was found to be significantly down-regulated by the vitamin D compounds. These results suggest that vitamin D compounds may serve as potential preventive agents to inhibit triple negative breast cancer by regulating cancer stem cells and differentiation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Calcitriol/analogs & derivatives , Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/drug therapy , Vitamins/pharmacology , Anticarcinogenic Agents/chemistry , Calcitriol/chemistry , Calcitriol/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/pathology , Vitamins/chemistry
10.
J Ginseng Res ; 40(4): 423-430, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27746696

ABSTRACT

BACKGROUND: The induction of cellular defensive genes such as phase II detoxifying and antioxidant enzymes is a highly effective strategy for protection against carcinogenesis as well as slowing cancer development. Transcription factor Nrf2 (nuclear factor E2-related factor 2) is responsible for activation of phase II enzymes induced by natural chemopreventive compounds. METHODS: Red ginseng oil (RGO) was extracted using a supercritical CO2 extraction system and chemical profile of RGO was investigated by GC/MS. Effects of RGO on regulation of the Nrf2/antioxidant response element (ARE) pathway were determined by ARE-luciferase assay, western blotting, and confocal microscopy. RESULTS: The predominant components of RGO were 9,12-octadecadienoic acid (31.48%), bicyclo[10.1.0]tridec-1-ene (22.54%), and 22,23-dihydrostigmasterol (16.90%). RGO treatment significantly increased nuclear translocation of Nrf2 as well as ARE reporter gene activity, leading to upregulation of heme oxygenase-1 and NAD(P)H:quinone oxidoreductase 1. Phosphorylation of the upstream kinases such as apoptosis signal-regulating kinase (ASK)1, mitogen-activated protein kinase (MAPK) kinase (MKK)4/7, c-Jun N-terminal kinase (JNK), and p38 MAPK were enhanced by treatment with RGO. In addition, RGO-mediated Nrf2 expression and nuclear translocation was attenuated by JNK inhibitor SP600125 and p38 MAPK inhibitor SB202190. CONCLUSION: RGO could be used as a potential chemopreventive agent, possibly by induction of Nrf2/ARE-mediated phase II enzymes via ASK1-MKK4/7-JNK and p38 MAPK signaling pathways.

11.
Int J Mol Sci ; 17(5)2016 May 18.
Article in English | MEDLINE | ID: mdl-27213339

ABSTRACT

In the present study, we characterized the antioxidant and hepatoprotective mechanisms underlying of wild grape seed procyanidins (WGP) against oxidative stress damage in ethanol-treated HepG2 cell and Sprague-Dawley (SD)-rat models. In HepG2 cells, WGP not only diminished the ethanol (EtOH, 100 mM)-induced reactive oxygen species (ROS) formation and cytochrome P450 2E1 (CYP2E1) expression, but also renovated both the activity and expression of antioxidant enzymes including catalase, superoxide dismutase, and glutathione peroxidase. Additionally, to investigate the hepatoprotective effect of WGP, rats were orally administered 10 or 50 mg/kg WGP once daily for seven days prior to the single oral administration of EtOH (6 g/kg). The results show that WGP administration decreased the EtOH-induced augment of the levels of serum aspartate transaminase and alanine transaminase as well as serum alcohol and acetaldehyde. WGP treatment upregulated the activities and protein levels of hepatic alcohol dehydrogenase, aldehyde dehydrogenase, and antioxidant enzymes but downregulated the protein expression level of liver CYP2E1 in EtOH-treated rats. Moreover, the decreased phosphorylation levels of mitogen activated protein kinases (MAPKs) by ethanol were induced in both HepG2 cell and rat models. Overall, pretreatment of WGP displayed the protective activity against EtOH-mediated toxicity through the regulation of antioxidant enzymes and alcohol metabolism systems via MAPKs pathways.


Subject(s)
Antioxidants/administration & dosage , Ethanol/toxicity , Liver Diseases, Alcoholic/prevention & control , Proanthocyanidins/administration & dosage , Vitis/chemistry , Alanine Transaminase/metabolism , Animals , Antioxidants/pharmacology , Aspartate Aminotransferases/metabolism , Cell Survival/drug effects , Gene Expression Regulation , Hep G2 Cells , Humans , Liver Diseases, Alcoholic/metabolism , MAP Kinase Signaling System/drug effects , Plant Extracts/chemistry , Plant Extracts/pharmacology , Proanthocyanidins/pharmacology , Rats , Rats, Sprague-Dawley
12.
Semin Cancer Biol ; 40-41: 170-191, 2016 10.
Article in English | MEDLINE | ID: mdl-27016037

ABSTRACT

Estrogen receptor (ER)-positive breast cancer, including luminal-A and -B, is the most common type of breast cancer. Extended exposure to estrogen is associated with an increased risk of breast cancer. Both ER-dependent and ER-independent mechanisms have been implicated in estrogen-mediated carcinogenesis. The ER-dependent pathway involves cell growth and proliferation triggered by the binding of estrogen to the ER. The ER-independent mechanisms depend on the metabolism of estrogen to generate genotoxic metabolites, free radicals and reactive oxygen species to induce breast cancer. A better understanding of the mechanisms that drive ER-positive breast cancer will help optimize targeted approaches to prevent or treat breast cancer. A growing emphasis is being placed on alternative medicine and dietary approaches toward the prevention and treatment of breast cancer. Many natural products and bioactive compounds found in foods have been shown to inhibit breast carcinogenesis via inhibition of estrogen induced oxidative stress as well as ER signaling. This review summarizes the role of bioactive natural products that are involved in the prevention and treatment of estrogen-related and ER-positive breast cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/prevention & control , Plant Extracts/pharmacology , Receptors, Estrogen/metabolism , Animals , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Diet , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic , Humans , Plant Extracts/therapeutic use , Signal Transduction
13.
Cancer Prev Res (Phila) ; 8(9): 807-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26130252

ABSTRACT

This study evaluated the anticancer activity and mechanism of action of a γ-tocopherol-rich tocopherol mixture, γ-TmT, in two different animal models of estrogen-induced breast cancer. The chemopreventive effect of γ-TmT at early (6 weeks), intermediate (18 weeks), and late (31 weeks) stages of mammary tumorigenesis was determined using the August-Copenhagen Irish rat model. Female rats receiving 17ß-estradiol (E2) implants were administered with different doses (0%, 0.05%, 0.1%, 0.3%, and 0.5%) of γ-TmT diet. Treatment with 0.3% and 0.5% γ-TmT decreased tumor volume and multiplicity. At 31 weeks, serum concentrations of E2 were significantly decreased by γ-TmT. γ-TmT preferentially induced expression of the E2-metabolizing enzyme CYP1A1, over CYP1B1 in the rat mammary tissues. Nrf2-dependent antioxidant response was stimulated by γ-TmT, as evident from enhanced expression of its downstream targets, NQO1, GCLM, and HMOX1. Serum concentrations of the oxidative stress marker, 8-isoprostane, were also decreased in the γ-TmT-treated groups. Treatment with γ-TmT increased expression of PPARγ and its downstream genes, PTEN and p27, whereas the cell proliferation marker, PCNA, was significantly reduced in γ-TmT-treated mammary tumors. In an orthotopic model in which human MCF-7 breast cancer cells were injected into the mammary fat pad of immunodeficient mice, γ-TmT inhibited E2-dependent tumor growth at all the doses tested. In conclusion, γ-TmT reduced mammary tumor development, in part through decreased E2 availability and reduced oxidative stress in mammary tissues; γ-TmT could thus be an effective agent for the prevention and treatment of E2-induced breast cancer.


Subject(s)
Antioxidants/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Estrogens/metabolism , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , gamma-Tocopherol/therapeutic use , Animals , Cell Line, Tumor , Dinoprost/analogs & derivatives , Dinoprost/chemistry , Estradiol/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Immunohistochemistry , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Stress , RNA, Messenger/metabolism , Rats , Time Factors
14.
Int J Mol Med ; 34(6): 1516-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25319719

ABSTRACT

Fermented black ginseng (FBG) is prepared by repeated steaming and drying processes with fresh ginseng followed by fermentation with Saccharomyces cerevisiae. It has recently been shown to have several bioactivities. FBG contains crude saponin (1,440 µg/ml), ginsenoside Rg2 (2.86 µg/ml), ginsenoside Rg3 (24.52 µg/ml), ginsenoside Rh1 (12.64 µg/ml), ginsenoside Rh2 (0.63 µg/ml) and ginsenoside Rf (1.32 µg/ml). The present study investigated the antioxidant defense properties of FBG against hydrogen peroxide (H2O2)-mediated oxidative stress in HepG2 human hepatocellular carcinoma cells. The increased production of reactive oxygen species (ROS) induced by H2O2 was attenuated in a dose-dependent manner when the cells were pre-treated with FBG (10-50 µg/ml). FBG induced both the expression and activity of antioxidant enzymes, such as superoxide dismutase, catalase and glutathione peroxidase in the H2O2-treated HepG2 cells. The inhibitory effects of FBG on the phosphorylation of upstream mitogen-activated protein kinases (MAPKs), such as c-Jun N-terminal kinase, extracellular signal-regulated kinase and p38 were also observed. Overall, our results demonstrate that FBG protects HepG2 cells from oxidative stress through the induction of antioxidant enzyme activity and the inhibition of MAPK pathways.


Subject(s)
Ginsenosides/pharmacology , Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Panax/chemistry , Saponins/pharmacology , Antioxidants/metabolism , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Catalase/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fermentation , Glutathione Peroxidase/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , Oxidants/pharmacology , Panax/metabolism , Phosphorylation/drug effects , Plant Preparations/chemistry , Plant Preparations/pharmacology , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
15.
J Agric Food Chem ; 62(34): 8668-78, 2014 Aug 27.
Article in English | MEDLINE | ID: mdl-25119859

ABSTRACT

Procyanidins, polymeric flavan-3-ols, are known to possess antioxidant, antiatherogenic, and anticarcinogenic properties. In the present study, we investigated the role of almond (Prunus amygdalus) skin procyanidins (ASP) in regulating the protein expression of phase II detoxifying and antioxidant enzymes in HepG2 cells and acetaminophen (APAP)-treated hepatotoxic mice. Treatments of ASP significantly induced the expression of phase II enzymes including NAD(P)H: quinoneoxidoreductase 1, catalase, glutathione peroxidase, and superoxide dismutase in the cells and mice. ASP also potently enhanced the expression of nuclear factor-E2-related factor 2 (Nrf2) and antioxidant response element (ARE)-reporter gene activity in vitro. APAP-induced hepatotoxic markers including AST and ALT in mice were inhibited by ASP administration. However, regulation of upstream kinases by ASP was different between in vitro and in vivo models. Collectively, ASP could induce the activation of Nrf2/ARE-mediated phase II detoxifying/antioxidant enzymes but with differential regulation on upstream kinases between in vitro and in vivo.


Subject(s)
Antioxidants/pharmacology , Biflavonoids/administration & dosage , Catechin/administration & dosage , Liver Diseases/prevention & control , Plant Extracts/administration & dosage , Proanthocyanidins/administration & dosage , Prunus/chemistry , Seeds/chemistry , Animals , Antioxidant Response Elements/drug effects , Catalase/genetics , Catalase/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Diseases/metabolism , Male , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Protective Agents/administration & dosage , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
16.
J Ginseng Res ; 38(1): 78-81, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24558315

ABSTRACT

The single oral administration of red ginseng oil (5000 mg/kg) to Sprague-Dawley rats induced no changes in behavioral patterns, clinical signs, and body weight, and hepatotoxicity parameters such as aspartate aminotransferase and alanine aminotransferase for 14 d. Therefore, these results suggest that the red ginseng oil is safe and nontoxic acutely.

17.
Oxid Med Cell Longev ; 2013: 409321, 2013.
Article in English | MEDLINE | ID: mdl-24260615

ABSTRACT

In the present study, the anti-inflammatory effect and underlying mechanisms of wild grape seeds procyanidins (WGP) were examined using lipopolysaccharide- (LPS-) stimulated RAW 264.7 cells. We used nitric oxide (NO) and prostaglandin E2 (PGE2) and reactive oxygen species (ROS) assays to examine inhibitory effect of WGP and further investigated the mechanisms of WGP suppressed LPS-mediated genes and upstream expression by Western blot and confocal microscopy analysis. Our data indicate that WGP significantly reduced NO, PGE2, and ROS production and also inhibited the expression of proinflammatory mediators such as inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) protein expressions. Consistently, WGP significantly reduced LPS-stimulated expression of proinflammatory cytokines such as tumor necrosis factor α (TNF-α) and interleukin- (IL-) 1 ß . Moreover, WGP prevented nuclear translocation of nuclear factor- κ B (NF κ B) p65 subunit by reducing inhibitory κ B- α (I κ B α) and NF κ B phosphorylation. Furthermore, we found that WGP inhibited LPS-induced phosphorylation of p38 mitogen-activated protein kinase (MAPK). Taken together, our results demonstrated that WGP exerts potent anti-inflammatory activity through the inhibition of iNOS and COX-2 by regulating NF κ B and p38 MAPK pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Grape Seed Extract/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Proanthocyanidins/pharmacology , Vitis/chemistry , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , I-kappa B Proteins/metabolism , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/enzymology , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , NF-KappaB Inhibitor alpha , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Molecules ; 17(12): 13769-86, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23174895

ABSTRACT

In this study, we investigated the anti-inflammatory effects of red ginseng marc oil (RMO) in the RAW 264.7 macrophage cell line. RMO was prepared by a supercritical CO(2) extraction of waste product generated after hot water extraction of red ginseng. RMO significantly inhibited the production of oxidative stress molecules such as nitric oxide and reactive oxygen species in lipopolysaccharide (LPS)-activated RAW 264.7 cells. Levels of inflammatory targets including prostaglandin E2, tumor necrosis factor-α, interleukin (IL)-1β and IL-6 were also reduced after the treatment with RMO. In addition, RMO diminished the expressions of inducible nitric oxide synthase and cyclooxygenase 2 at both mRNA and protein levels. Blockade of nuclear translocation of the p65 subunit of nuclear factor κB (NFκB) was also observed after the treatment of RMO. Furthermore, RMO decreased the phosphorylations of p38 mitogen-activated protein kinase (MAPK) and its upstream kinases including MAPK kinases 3/6 (MKK3/6) and TAK 1 (TGF-β activated kinase 1). Gas chromatographic analysis on RMO revealed that RMO contained about 10% phytosterols including sitosterol, stigmasterol and campesterol which may contribute to the anti-inflammatory properties of RMO. Taken together, these results suggest that the anti-inflammatory effect of RMO in LPS-induced RAW 264.7 macrophages could be associated with the inhibition of NFκB transcriptional activity, possibly via blocking the p38 MAPK pathway.


Subject(s)
Anti-Inflammatory Agents , Inflammation , Macrophages/metabolism , Panax , Plant Oils/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Panax/chemistry , Phosphorylation , Plant Oils/chemistry , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
19.
Molecules ; 17(7): 8037-55, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22763741

ABSTRACT

The rhizome of ginger (Zingiber officinale Roscoe) is known to have several bioactive compounds including gingerols and shogaols which possess beneficial health properties such as anti-inflammatory and chemopreventive effects. Based on recent observations that 6-shogaol may have more potent bioactivity than 6-gingerol, we obtained a 6-shogaol-rich extract from ginger and examined its effects on the nuclear factor E2-related factor2 (Nrf2)/antioxidant response element (ARE) pathway in vitro and in vivo. 6-Shogaol-rich extract was produced by extracting ginger powder with 95% ethanol at 80 °C after drying at 80 °C (GEE8080). GEE8080 contained over 6-fold more 6-shogaol compared to the room temperature extract (GEE80RT). In HepG2 cells, GEE8080 displayed much stronger inductions of ARE-reporter gene activity and Nrf2 expression than GEE80RT. GEE8080 stimulated phosphorylations of mitogen-activated protein kinases (MAPKs) such as ERK, JNK, and p38. Moreover, the GEE8080-induced expressions of Nrf2 and HO-1 were attenuated by treatments of SB202190 (a p38 specific inhibitor) and LY294002 (an Akt specific inhibitor). In a mouse model, the GEE8080 decreased the diethylnitrosamine (DEN)-mediated elevations of serum aspartate transaminase and alanine transaminase as well as the DEN-induced hepatic lipid peroxidation. Inductions of Nrf2 and HO-1 by GEE8080 were also confirmed in the mice. In addition, the administration of GEE8080 to the mice also restored the DEN-reduced activity and protein expression of hepatic antioxidant enzymes such as superoxide dismutase, glutathione peroxidase and catalase. In conclusion, GEE8080, a 6-shogaol-rich ginger extract, may enhance antioxidant defense mechanism through the induction of Nrf2 and HO-1 regulated by p38 MAPK and PI3k/Akt pathway in vitro and in vivo.


Subject(s)
Antioxidants/metabolism , Catechols/pharmacology , Plant Extracts/pharmacology , Up-Regulation/drug effects , Zingiber officinale/chemistry , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Diethylnitrosamine , Genes, Reporter/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Liver/drug effects , Liver/enzymology , Luciferases/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Plant Extracts/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Response Elements/genetics , Thiobarbituric Acid Reactive Substances/metabolism
20.
Int J Mol Sci ; 13(2): 2314-2330, 2012.
Article in English | MEDLINE | ID: mdl-22408456

ABSTRACT

The aim of this study was to evaluate the antioxidant mechanisms of red ginseng essential oil (REO) in cells as well as in an animal model. REO was prepared by a supercritical CO(2) extraction of waste-products generated after hot water extraction of red ginseng. In HepG2 cells, REO diminished the H(2)O(2)-mediated oxidative stress and also restored both the activity and expression of antioxidant enzymes such as superoxide dismutase, catalase and glutathione peroxidase. Administration of REO inhibited the phosphorylation of upstream mitogen-activated protein kinases (MAPKs) such as c-Jun N-terminal kinase, extracellular signal-regulated kinase, and p38. In mice, the CCl(4)-mediated elevation of serum aspartate transaminase and alanine transaminase as well as the induction of hepatic lipid peroxidation were decreased by REO administration. REO treatments also resulted in up-regulation of the antioxidant enzyme expression in the liver. Moreover, increased phosphorylations of MAPKs were inhibited after REO administration. Overall, REO seems to protect the liver from oxidative stress through the activation and induction of antioxidant enzymes via inhibition of MAPKs pathways.


Subject(s)
Antioxidants/pharmacology , Carbon Tetrachloride/toxicity , Cytoprotection/drug effects , Hydrogen Peroxide/toxicity , Liver/drug effects , Oils, Volatile/pharmacology , Panax/chemistry , Animals , Carbon Tetrachloride Poisoning/prevention & control , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Plant Extracts/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...