Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Mol Nutr Food Res ; 65(20): e2100539, 2021 10.
Article in English | MEDLINE | ID: mdl-34406707

ABSTRACT

SCOPE: This study investigates the mechanism of action and functional effects of coffee extracts in colonic cells, on intestinal stem cell growth, and inhibition of dextran sodium sulfate (DSS)-induced intestinal barrier damage in mice. METHODS AND RESULTS: Aqueous coffee extracts induced Ah receptor (AhR) -responsive CYP1A1, CYP1B1, and UGT1A1 gene expression in colon-derived Caco2 and YAMC cells. Tissue-specific AhR knockout (AhRf/f x Lgr5-GFP-CreERT2 x Villin-Cre), wild-type (Lgr5-CreERT2 x Villin-Cre) mice are sources of stem cell enriched organoids and both coffee extracts and norharman, an AhR-active component of these extracts inhibited stem cell growth. Coffee extracts also inhibit DSS-induced damage to intestinal barrier function and DSS-induced mucosal inflammatory genes such as IL-6 and TGF-ß1 in wild-type (AhR+/+ ) but not AhR-/- mice. In contrast, coffee does not exhibit protective effects in intestinal-specific AhR knockout mice. Coffee extracts also enhanced overall formation of AhR-active microbial metabolites. CONCLUSIONS: In colon-derived cells and in the mouse intestine, coffee induced several AhR-dependent responses including gene expression, inhibition of intestinal stem cell-enriched organoid growth, and inhibition of DSS-induced intestinal barrier damage. We conclude that the anti-inflammatory effects of coffee in the intestine are due, in part, to activation of AhR signaling.


Subject(s)
Coffee , Colon/drug effects , Plant Extracts/pharmacology , Receptors, Aryl Hydrocarbon/physiology , Animals , Caco-2 Cells , Colon/metabolism , Cytochrome P-450 CYP1A1/physiology , Cytochrome P-450 CYP1B1/physiology , Dextran Sulfate/toxicity , Female , Humans , Male , Mice
3.
PLoS One ; 15(2): e0228436, 2020.
Article in English | MEDLINE | ID: mdl-32027669

ABSTRACT

BACKGROUND: Cytochrome P450 1b1 (Cyp1b1) deletion and dietary retinol deficiency during pregnancy (GVAD) affect perinatal liver functions regulated by Srebp. Cyp1b1 is not expressed in perinatal liver but appears in the E9.5 embryo, close to sites of retinoic acid (RA) signaling. HYPOTHESIS: Parallel effects of Cyp1b1 and retinol on postnatal Srebp derive from effects in the developing liver or systemic signaling. APPROACH: Cluster postnatal increases in hepatic genes in relation to effects of GVAD or Cyp1b1 deletion. Sort expression changes in relation to genes regulated by Srebp1 and Srebp2.Test these treatments on embryos at E9.5, examining changes at the site of liver initiation. Use in situ hybridization to resolve effects on mRNA distributions of Aldh1a2 and Cyp26a1 (RA homeostasis); Hoxb1 and Pax6 (RA targets). Assess mice lacking Lrat and Rbp4 (DKO mice) that severely limits retinol supply to embryos. RESULTS: At birth, GVAD and Cyp1b1 deletion stimulate gene markers of hepatic stellate cell (HSC) activation but also suppress Hamp. These treatments then selectively prevent the postnatal onset of genes that synthesize cholesterol (Hmgcr, Sqle) and fatty acids (Fasn, Scd1), but also direct cholesterol transport (Ldlr, Pcsk9, Stard4) and retinoid synthesis (Aldh1a1, Rdh11). Extensive support by Cyp1b1 is implicated, but with distinct GVAD interventions for Srebp1 and Srebp2. At E9.5, Cyp1b1 is expressed in the septum transversum mesenchyme (STM) with ß-carotene oxygenase (Bco1) that generates retinaldehyde. STM provides progenitors for the HSC and supports liver expansion. GVAD and Cyp1b1-/- do not affect RA-dependent Hoxb1 and Pax6. In DKO embryos, RA-dependent Cyp26a1 is lost but Hoxb1 is sustained with Cyp1b1 at multiple sites. CONCLUSION: Cyp1b1-/- suppresses genes supported by Srebp. GVAD effects distinguish Srebp1 and Srebp2 mediation. Srebp regulation overlaps appreciably in cholesterol and retinoid homeostasis. Bco1/Cyp1b1 partnership in the STM may contribute to this later liver regulation.


Subject(s)
Cholesterol/biosynthesis , Cytochrome P-450 CYP1B1/physiology , Fetal Development , Liver/metabolism , Sterol Regulatory Element Binding Proteins/physiology , Tretinoin/metabolism , Animals , Animals, Newborn , Cytochrome P-450 CYP1B1/genetics , Embryo, Mammalian , Female , Fetal Development/drug effects , Fetal Development/genetics , Liver/drug effects , Liver/embryology , Liver/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Retinol-Binding Proteins, Plasma/genetics , Retinol-Binding Proteins, Plasma/metabolism , Tretinoin/pharmacology
4.
J Toxicol Environ Health A ; 81(9): 278-287, 2018.
Article in English | MEDLINE | ID: mdl-29473798

ABSTRACT

Cytochrome P450 1B1 (CYP1B1), a well-known oncogene, has garnered wide attention because of its tumor-specific expression pattern and actions as a carcinogenic factor. Although CYP1B1 might play a crucial role in carcinogenesis, the detailed molecular mechanisms underlying oncogenic involvement in cancer development remain unclear. The present study investigated the manner in which CYP1B1 promotes survival of various cancer cells. Treatment with 2,2',4,6'-tetramethoxystilbene (TMS), a specific CYP1B1 inhibitor, significantly inhibited cell viability in human breast cancer and leukemia cell lines, including MCF-7, MDA-MB-231, HL-60, and U937 cells. In order to characterize the cellular functions of CYP1B1 associated with cancer cell survival, the relationship between this oncogene and death receptor 4 (DR4) was determined. Following induction or inhibition of CYP1B1, mRNA and protein expression levels of DR4 were measured, and this oncogene was found to significantly repress DR4 mRNA and protein expression. Further, the suppression of DR4 by CYP1B1 was restored with 5-aza-2'-deoxycytidine (5-aza-dC), a DNA methyltransferase inhibitor, indicating that DNA methylation may be involved in CYP1B1-mediated DR4 inhibition. Methylation-specific polymerase chain reaction (PCR) in CYP1B1-overexpressed HL-60 cells revealed that this oncogene induced hypermethylation on DR4 promoter. Interestingly, data showed that DR4 suppression of CYP1B1 is mediated by the DNA-binding ability of specificity protein 1 (Sp1). These findings suggest that CYP1B1 promotes cancer cell survival through involvement of DNA methylation-mediated DR4 inhibition and that Sp1 may act as key mediator required for oncogenic action.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Cytochrome P-450 CYP1B1/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Sp1 Transcription Factor/genetics , Cell Line, Tumor , Cytochrome P-450 CYP1B1/genetics , DNA Methylation/drug effects , HL-60 Cells , Humans , MCF-7 Cells , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Sp1 Transcription Factor/metabolism , U937 Cells
5.
PLoS One ; 11(3): e0151598, 2016.
Article in English | MEDLINE | ID: mdl-26981862

ABSTRACT

Cytochrome P450 1B1 (CYP1B1) is a major E2 hydroxylase involved in the metabolism of potential carcinogens. CYP1B1 expression has been reported to be higher in tumors compared to normal tissues, especially in hormone-related cancers including breast, ovary, and prostate tumors. To explore the role of CYP1B1 in cancer progression, we investigated the action of CYP1B1 in cells with increased CYP1B1 via the inducer 7,12-dimethylbenz[α]anthracene (DMBA) or an overexpression vector, in addition to decreased CYP1B1 via the inhibitor tetramethoxystilbene (TMS) or siRNA knockdown. We observed that CYP1B1 promoted cell proliferation, migration, and invasion in MCF-7 and MCF-10A cells. To understand its molecular mechanism, we measured key oncogenic proteins including ß-catenin, c-Myc, ZEB2, and matrix metalloproteinases following CYP1B1 modulation. CYP1B1 induced epithelial-mesenchymal transition (EMT) and activated Wnt/ß-catenin signaling via upregulation of CTNNB1, ZEB2, SNAI1, and TWIST1. Sp1, a transcription factor involved in cell growth and metastasis, was positively regulated by CYP1B1, and suppression of Sp1 expression by siRNA or DNA binding activity using mithramycin A blocked oncogenic transformation by CYP1B1. Therefore, we suggest that Sp1 acts as a key mediator for CYP1B1 action. Treatment with 4-hydroxyestradiol (4-OHE2), a major metabolite generated by CYP1B1, showed similar effects as CYP1B1 overexpression, indicating that CYP1B1 activity mediated various oncogenic events in cells. In conclusion, our data suggests that CYP1B1 promotes cell proliferation and metastasis by inducing EMT and Wnt/ß-catenin signaling via Sp1 induction.


Subject(s)
Cell Proliferation/drug effects , Cytochrome P-450 CYP1B1/physiology , Epithelial-Mesenchymal Transition , Neoplasm Metastasis , Signal Transduction , Sp1 Transcription Factor/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/biosynthesis , Cytochrome P-450 CYP1B1/genetics , Estrogens, Catechol/metabolism , Humans , Up-Regulation
6.
Blood ; 127(18): 2231-40, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26966088

ABSTRACT

The mechanisms regulating megakaryopoiesis and platelet production (thrombopoiesis) are still incompletely understood. Identification of a progenitor with enhanced thrombopoietic capacity would be useful to decipher these mechanisms and to improve our capacity to produce platelets in vitro. Differentiation of peripheral blood CD34(+) cells in the presence of bone marrow-human mesenchymal stromal cells (MSCs) enhanced the production of proplatelet-bearing megakaryocytes (MKs) and platelet-like elements. This was accompanied by enrichment in a MK precursor population exhibiting an intermediate level of CD41 positivity while maintaining its expression of CD34. Following sorting and subculture with MSCs, this CD34(+)CD41(low) population was able to efficiently generate proplatelet-bearing MKs and platelet-like particles. Similarly, StemRegenin 1 (SR1), an antagonist of the aryl hydrocarbon receptor (AhR) transcription factor known to maintain CD34 expression of progenitor cells, led to an enriched CD34(+)CD41(low) fraction and to an increased capacity to generate proplatelet-producing MKs and platelet-like elements ultrastructurally and functionally similar to circulating platelets. The effect of MSCs, like that of SR1, appeared to be mediated by an AhR-dependent mechanism because both culture conditions resulted in repression of its downstream effector CYP1B1. This newly described isolation of a precursor exhibiting strong MK potential could be exploited to study normal and abnormal thrombopoiesis and for in vitro platelet production.


Subject(s)
Megakaryocyte Progenitor Cells/cytology , Receptors, Aryl Hydrocarbon/physiology , Thrombopoiesis/physiology , Antigens, CD34/analysis , Blood Platelets/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free , Cytochrome P-450 CYP1B1/physiology , Humans , Immunophenotyping , Platelet Count , Platelet Membrane Glycoprotein IIb/analysis , Purines/pharmacology , Signal Transduction
7.
Toxicol Lett ; 230(2): 304-13, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-23994263

ABSTRACT

Eupatorin-5-methyl ether (E5M) is a flavone containing 4 methoxy groups that is present in plants with medicinal activity, whereas luteolin (L) is a polyhydroxylated flavone commonly encountered in dietary products. In the present study we investigated the interaction of the two flavonoids with cytochrome P450 CYP1 enzymes in breast cancer MCF7 cells. Both compounds induced a dose dependent increase in CYP1A1 and CYP1B1 mRNA levels, as well as in EROD activity, a marker of CYP1 enzyme activity. Induction of cytochrome P450 CYP1 expression by E5M was accompanied by translocation of the ligand-activated transcription factor AhR to the nucleus, as demonstrated by confocal immunofluoresence. More importantly, although E5M was less active than L in inhibiting proliferation of MCF7 cells, when the cells were pretreated with the CYP1 inducer Benzo[a]pyrene (BaP) the potency of E5M was augmented. HPLC and LC-MS analysis revealed that E5M was metabolized to a major conversion product assigned E5M1 resulting from one step demethylation reaction in MCF7 cells whereas L metabolism by recombinant CYP1A1 did not reveal any metabolites. E5M1 production in BaP-induced MCF7 cells was attenuated in the presence of the CYP1A1 inhibitor α-napthoflavone. E5M further induced a dose dependent increase in the cell signaling proteins p21, JNK and p-JNK in MCF7 cells. This effect was enhanced in BaP pretreated cells and was associated with G1 arrest and a small percentage of apoptosis (3.5%). E5M antiproliferative effect in BaP pretreated cells was attenuated in the presence of the CYP1A1 inhibitor α-napthoflavone, as demonstrated by Western blotting and FACS analysis. Taken together the results demonstrate that BaP sensitizes MCF7 cells to E5M antiproliferative activity via enhanced induction of p21, JNK and p-JNK that in turn results by cytochrome P450 CYP1-mediated conversion to the metabolite E5M1.


Subject(s)
Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/physiology , Cytochrome P-450 CYP1B1/physiology , Flavones/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Female , Humans , MAP Kinase Kinase 4/metabolism , MCF-7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...