Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Biotechnol ; 21(1): 42, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34281556

ABSTRACT

BACKGROUND: Protein C receptor (Procr) has recently been shown to mark resident adult stem cells in the mammary gland, vascular system, and pancreatic islets. More so, high Procr expression was also detected and used as indicator for subsets of triple-negative breast cancers (TNBCs). Previous study has revealed Procr as a target of Wnt/ß-catenin signaling; however, direct upstream regulatory mechanism of Procr remains unknown. To comprehend the molecular role of Procr during physiology and pathology, elucidating the upstream effectors of Procr is necessary. Here, we provide a system for screening negative regulators of Procr, which could be adapted for broad molecular analysis on membrane proteins. RESULTS: We established a screening system which combines CRISPR-Cas9 guided gene disruption with fluorescence activated cell sorting technique (FACS). CommaDß (murine epithelial cells line) was used for the initial Procr upstream effector screening using lentiviral CRISPR-gRNA library. Shortlisted genes were further validated through individual lentiviral gRNA infection followed by Procr expression evaluation. Adam17 was identified as a specific negative inhibitor of Procr expression. In addition, MDA-MB-231 cells and Hs578T cells (human breast cancer cell lines) were used to verify the conserved regulation of ADAM17 over PROCR expression. CONCLUSION: We established an efficient CRISPR-Cas9/FACS screening system, which identifies the regulators of membrane proteins. Through this system, we identified Adam17 as the negative regulator of Procr membrane expression both in mammary epithelial cells and breast cancer cells.


Subject(s)
ADAM17 Protein/metabolism , Endothelial Protein C Receptor/genetics , Lentivirus/genetics , Mammary Glands, Human/enzymology , ADAM17 Protein/genetics , Base Sequence , Cell Line , Down-Regulation , Endothelial Protein C Receptor/metabolism , Gene Library , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Lentivirus/metabolism , RNA, Guide, Kinetoplastida/genetics
2.
Int J Cancer ; 145(3): 817-829, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30671946

ABSTRACT

The hotspot mutation H1047R in the oncogenic PIK3CA gene is frequently detected in breast cancer and enhances the enzymatic activity of PI3K to activate AKT/mTOR signaling cascade. Aberrant elevated PI3K activation has been reported to promote the tumorigenesis of breast cancer, but the mechanisms underlying are still needed to be elucidated. Here, we found that continuously activating PIK3CAH1047R conferred human mammary epithelial MCF-10A cells to cellular senescence upon serum-starvation. Similarly, breast cancer T47D and HCC1954 cells harboring H1047R mutation were senescent when cells were deprived of serum. PI3K/AKT/mTOR axis but not p53 or RB might be required for the induction of senescence. Notably, membrane metallo-endopeptidase (MME) was identified as a downstream effector of PI3K to mediate the induction of senescence, which might be associated with its glycosylation. Senescent cells elicited a distinct secretome dependent on PI3K and MME. Specifically, IL-6 promoted the proliferation of normal cells and CCL2 induced the M2-like polarization of macrophages, which might create an immunosuppressive microenvironment during the initiation and/or development of breast cancer. This study shed new light on the tumorigenesis induced by hyper-activated PI3K and might provide new clues for the prevention and therapy of breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Metalloendopeptidases/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cellular Senescence/physiology , Chemokine CCL2/metabolism , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Epithelial Cells/cytology , Epithelial Cells/enzymology , Glycosylation , Humans , Interleukin-6/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/enzymology , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
3.
Toxicol Mech Methods ; 29(9): 686-692, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31364906

ABSTRACT

Life-long estrogen exposure is one of the major risk factors in the development and progression of breast cancer. However, little is known about the molecular mechanisms, by which chronic exposure to estrogen contributes to breast carcinogenesis. The aim of the present study was to investigate the effects of long-term exposure with 4-hydroxyestradiol (4-OHE2) on acquired cancer characteristics of human mammary epithelial MCF-10A cells. The possible regulators were further studied in chronic 4-OHE2-treated MCF-10A cells. We observed that MCF-10A cells long-term exposed to 4-OHE2 acquire the characteristics of cancer cells, such as enhanced cell growth, EMT properties, and increased migration and invasiveness. Moreover, the expression of CYP1B1 was significantly elevated in long-term 4-OHE2-treated MCF-10A cells. Block of CYP1B1 significantly reduced the cancer cell characteristics in long-term 4-OHE2-treated MCF-10A cells. Our results indicated that 4-OHE2 mediated enhanced cancer cell characteristics in mammary epithelial cells are an important key event for breast carcinogenesis process. CYP1B1 partially contributes to the 4-OHE2 induced cancer cell characteristics in MCF-10A cells. Targeting CYP1B1 might offer a new strategy for the treatment of estrogen-induced breast cancer.


Subject(s)
Carcinogenesis/chemically induced , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytochrome P-450 CYP1B1/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Estrogens, Catechol/toxicity , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Culture Techniques , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/enzymology , Up-Regulation
4.
J Cell Physiol ; 233(3): 2464-2475, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28771726

ABSTRACT

Annexin A2 (AnxA2) has been shown to play multiple roles in growth, development, and metabolism, but the functions of AnxA2 and the signaling pathways associated with AnxA2 are still not fully understood. In this study, we aim to reveal whether and how AnxA2 could be involved in milk synthesis and proliferation of bovine mammary epithelial cells (BMECs). Using gene function study approaches, we found that AnxA2 positively regulates PIP3 level, phosphorylation of mTOR, and protein levels of SREBP-1c and Cyclin D1 leading to milk synthesis and cell proliferation. We further observed that both AnxA2-36 kD phosphorylated form and AnxA2-33 kD protein could be induced from AnxA2-36 kD protein in BMECs under methionine, leucine, estrogen or prolactin stimulation. These above results strongly demonstrate that AnxA2 functions as a critical regulator for amino acid or hormone-induced milk synthesis and cell proliferation via the PI3K-mTOR-SREBP-1c/Cyclin D1 signaling pathway.


Subject(s)
Annexin A2/metabolism , Cell Proliferation , Epithelial Cells/enzymology , Mammary Glands, Human/enzymology , Milk Proteins/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Animals , Annexin A2/genetics , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/metabolism , Epithelial Cells/drug effects , Estrogens/pharmacology , Female , Humans , Leucine/pharmacology , Mammary Glands, Human/drug effects , Methionine/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Progesterone/pharmacology , RNA Interference , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Transfection
5.
Biochem Biophys Res Commun ; 505(2): 385-391, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30262139

ABSTRACT

In vitro, the rat Fatty Acid Desaturase 3 (FADS3) gene was shown to code for an enzyme able to catalyze the unexpected Δ13-desaturation of trans-vaccenic acid, producing the trans11,cis13-conjugated linoleic acid (CLA) isomer. FADS3 may therefore be the first methyl-end trans-vaccenate Δ13-desaturase functionally characterized in mammals, but the proof of this concept is so far lacking in vivo. The present study therefore aimed at investigating further the putative in vivo synthesis of trans11,cis13-CLA from dietary trans-vaccenic acid in rodents. During one week of pregnancy and two weeks post-partum, Sprague-Dawley female rats were fed two diets either high (10.0% of fatty acids and 3.8% of energy intake) or low (0.4% of fatty acids and 0.2% of energy intake) in trans-vaccenic acid. The trans11,cis13-CLA was specifically detected, formally identified and reproducibly quantified (0.06% of total fatty acids) in the mammary gland phospholipids of lactating female rats fed the high trans-vaccenic acid-enriched diet. This result was consistent with FADS3 mRNA expression being significantly higher in the lactating mammary gland than in the liver. Although the apparent metabolic conversion is low, this physiological evidence demonstrates the existence of this new pathway described in the lactating mammary gland and establishes the FADS3 enzyme as a reliable mammalian trans-vaccenate Δ13-desaturase in vivo.


Subject(s)
Fatty Acid Desaturases/metabolism , Linoleic Acids, Conjugated/metabolism , Mammary Glands, Human/metabolism , Oleic Acids/metabolism , Animals , Catalysis , Diet, Fat-Restricted , Diet, High-Fat , Fatty Acid Desaturases/genetics , Female , Humans , Lactation , Linoleic Acids, Conjugated/biosynthesis , Mammary Glands, Human/enzymology , RNA, Messenger/metabolism , Rats
6.
J Biol Chem ; 291(3): 1529-37, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26589799

ABSTRACT

MicroRNA regulation of protein expression plays an important role in mediating many cellular processes, from cell proliferation to cell death. The human microRNA miR-424 is up-regulated in response to anti-proliferative cytokines, such as transforming growth factor ß (TGFß), and directly represses cell cycle progression. Our laboratory recently established that microRNA can be used as a proxy to identify biological roles of glycosylation enzymes (glycogenes). Herein we identify MGAT4A, OGT, and GALNT13 as targets of miR-424. We demonstrate that MGAT4A, an N-acetylglucosaminyltransferase that installs the ß-1,4 branch of N-glycans, is directly regulated by miR-424 in multiple mammary epithelial cell lines and observe the loss of MGAT4A in response to TGFß, an inducer of miR-424. Knockdown of MGAT4A induces cell cycle arrest through decreasing CCND1 levels. MGAT4A does not affect levels of ß-1,6 branched N-glycans, arguing that this effect is specific to ß-1,4 branching and not due to gross changes in overall N-linked glycosylation. This work provides insight into the regulation of cell cycle progression by specific N-glycan branching patterns.


Subject(s)
Mammary Glands, Human/metabolism , MicroRNAs/metabolism , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/antagonists & inhibitors , RNA Interference , Cell Cycle , Cell Line , Cell Proliferation , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Enzyme Repression , Genes, Reporter , Glycosylation , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/enzymology , MicroRNAs/agonists , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , RNA, Small Interfering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transforming Growth Factor beta/metabolism
7.
Molecules ; 22(6)2017 Jun 10.
Article in English | MEDLINE | ID: mdl-28604588

ABSTRACT

Docosahexaenoic acid (DHA), an ω-3 fatty acid abundant in fish oils, has diverse health beneficial effects, such as anti-oxidative, anti-inflammatory, neuroprotective, and chemopreventive activities. In this study, we found that DHA induced expression of two representative antioxidant/cytoprotective enzymes, heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase (NQO1), in human mammary epithealial (MCF-10A) cells. DHA-induced upregulation of these enzymes was accompanied by enhanced translocation of the redox-sensitive transcription factor Nrf2 into the nucleus and its binding to antioxidant response element. Nrf2 gene silencing by siRNA abolished the DHA-induced expression of HO-1 and NQO1 proteins. When MCF-10A cells were transfected with mutant constructs in which the cysteine 151 or 288 residue of Keap1 was replaced by serine, DHA-induced expression of HO-1 and NQO1 was markedly reduced. Moreover, DHA activated protein kinase C (PKC)δ and induced Nrf2 phosphorylation. DHA-induced phosphorylation of Nrf2 was abrogated by the pharmacological PKCδ inhibitor rottlerin or siRNA knockdown of its gene expression. The antioxidants N-acetyl-l-cysteine and Trolox attenuated DHA-induced activation of PKCδ, phosphorylation of Nrf2, and and its target protein expression. In conclusion, DHA activates Nrf2, possibly through modification of critical Keap1 cysteine 288 residue and PKCδ-mediated phosphorylation of Nrf2, leading to upregulation of HO-1 and NQO1 expression.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Heme Oxygenase-1/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , Acetylcysteine/administration & dosage , Antioxidants/administration & dosage , Chromans/administration & dosage , Gene Expression Regulation, Enzymologic/drug effects , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Mammary Glands, Human/drug effects , Mammary Glands, Human/enzymology , Phosphorylation/drug effects , Protein Kinase C-delta/genetics , RNA, Small Interfering/genetics
8.
FASEB J ; 29(9): 3990-4000, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26071407

ABSTRACT

Compared to normal tissues, many cancer cells overexpress autotaxin (ATX). This secreted enzyme produces extracellular lysophosphatidate, which signals through 6 GPCRs to drive cancer progression. Our previous work showed that ATX inhibition decreases 4T1 breast tumor growth in BALB/c mice by 60% for about 11 d. However, 4T1 cells do not produce significant ATX. Instead, the ATX is produced by adjacent mammary adipose tissue. We investigated the molecular basis of this interaction in human and mouse breast tumors. Inflammatory mediators secreted by breast cancer cells increased ATX production in adipose tissue. The increased lysophosphatidate signaling further increased inflammatory mediator production in adipose tissue and tumors. Blocking ATX activity in mice bearing 4T1 tumors with 10 mg/kg/d ONO-8430506 (a competitive ATX inhibitor, IC90 = 100 nM; Ono Pharma Co., Ltd., Osaka, Japan) broke this vicious inflammatory cycle by decreasing 20 inflammatory mediators by 1.5-8-fold in cancer-inflamed adipose tissue. There was no significant decrease in inflammatory mediator levels in fat pads that did not bear tumors. ONO-8430506 also decreased plasma TNF-α and G-CSF cytokine levels by >70% and leukocyte infiltration in breast tumors and adjacent adipose tissue by >50%. Hence, blocking tumor-driven inflammation by ATX inhibition is effective in decreasing tumor growth in breast cancers where the cancer cells express negligible ATX.


Subject(s)
Adipose Tissue/enzymology , Breast Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/enzymology , Mammary Glands, Human/enzymology , Mammary Neoplasms, Experimental/enzymology , Neoplasm Proteins/biosynthesis , Phosphoric Diester Hydrolases/biosynthesis , Adipose Tissue/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Phosphoric Diester Hydrolases/genetics
9.
J Biol Chem ; 288(52): 36926-35, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24217252

ABSTRACT

We investigated the role of protein-tyrosine phosphatase α (PTPα) in regulating signaling by the ErbB2 oncoprotein in mammary epithelial cells. Using this model, we demonstrated that activation of ErbB2 led to the transient inactivation of PTPα, suggesting that attenuation of PTPα activity may contribute to enhanced ErbB2 signaling. Furthermore, RNAi-induced suppression of PTPα led to increased cell migration in an ErbB2-dependent manner. The ability of ErbB2 to increase cell motility in the absence of PTPα was characterized by prolonged interaction of GRB7 with ErbB2 and increased association of ErbB2 with a ß1-integrin-rich complex, which depended on GRB7-SH2 domain interactions. Finally, suppression of PTPα resulted in increased phosphorylation of focal adhesion kinase on Tyr-407, which induced the recruitment of vinculin and the formation of a novel focal adhesion kinase complex in response to ErbB2 activation in mammary epithelial cells. Collectively, these results reveal a new role for PTPα in the regulation of motility of mammary epithelial cells in response to ErbB2 activation.


Subject(s)
Cell Movement/physiology , Epithelial Cells/enzymology , Focal Adhesion Kinase 1/metabolism , Mammary Glands, Human/enzymology , Receptor, ErbB-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Cell Line , Epithelial Cells/cytology , Focal Adhesion Kinase 1/genetics , Humans , Male , Mammary Glands, Human/cytology , Phosphorylation/physiology , Receptor, ErbB-2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Signal Transduction/physiology , Vinculin/genetics , Vinculin/metabolism
10.
Mol Biol Rep ; 41(1): 19-24, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24253898

ABSTRACT

The objective of this study is to compare the expression level of MAP3K1 between normal mammary gland cells and breast cancer cells, and to analyze the effects of silencing MAP3K1 on breast cancer cells with paclitaxel treatment. Western blotting analysis was used to detect the expression level of MAP3K1 in MCF-7 and MCF-12F cells. The effect of gene silencing through different siRNAs was determined by realtime-PCR. MTT assay was used to test the cell proliferation. Cell cycle was detected by flow cytometry. MAP3K1 protein expression level in breast cancer cells was higher than that in normal mammary gland cells. MAP3K1 siRNA transfection significantly reduced the expression level of MAP3K1, and enhanced paclitaxel-induced cell proliferation inhibition and cell cycle arrest in breast cancer cells. Targeting MAP3K1 expression through small RNA interference can promote the therapeutic effects of paclitaxel in breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle Checkpoints/drug effects , MAP Kinase Kinase Kinase 1/genetics , Paclitaxel/pharmacology , Breast Neoplasms , Cell Proliferation , Cyclin B1/genetics , Cyclin B1/metabolism , Female , Gene Expression , Gene Knockdown Techniques , Humans , MAP Kinase Kinase Kinase 1/metabolism , MCF-7 Cells , Mammary Glands, Human/enzymology , Mammary Glands, Human/pathology , RNA Interference , RNA, Small Interfering/genetics , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
11.
Int J Cancer ; 132(9): 2200-8, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23024001

ABSTRACT

Inhibition of histone deacetylase (HDAC) activity as stand-alone or combination therapy represents a promising therapeutic approach in oncology. The pan- or class I HDAC inhibitors (HDACi) currently approved or in clinical studies for oncology give rise to dose-limiting toxicities, presumably because of the inhibition of several HDACs. This could potentially be overcome by selective blockade of single HDAC family members. Here we report that HDAC11, the most recently identified zinc-dependent HDAC, is overexpressed in several carcinomas as compared to corresponding healthy tissues. HDAC11 depletion is sufficient to cause cell death and to inhibit metabolic activity in HCT-116 colon, PC-3 prostate, MCF-7 breast and SK-OV-3 ovarian cancer cell lines. The antitumoral effect induced can be mimicked by enforced expression of a catalytically impaired HDAC11 variant, suggesting that inhibition of the enzymatic activity of HDAC11 by small molecules could trigger the desired phenotypic changes. HDAC11 depletion in normal cells causes no changes in metabolic activity and viability, strongly suggesting that tumor-selective effects can be achieved. Altogether, our data show that HDAC11 plays a critical role in cancer cell survival and may represent a novel drug target in oncology.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Mammary Glands, Human/enzymology , Neoplasms/pathology , Small Molecule Libraries , Blotting, Western , Caspase 3/metabolism , Cells, Cultured , Flow Cytometry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mammary Glands, Human/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
Stem Cells ; 30(2): 344-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22131125

ABSTRACT

Elevated aldehyde dehydrogenase (ALDH) expression/activity has been identified as an important biomarker of primitive cells in various normal and malignant human tissues. Here we examined the level and type of ALDH expression and activity in different subsets of phenotypically and functionally defined normal human mammary cells. We find that the most primitive human mammary stem and progenitor cell types with bilineage differentiation potential show low ALDH activity but undergo a marked, selective, and transient upregulation of ALDH activity at the point of commitment to the luminal lineage. This mirrors a corresponding change in transcripts and protein levels of ALDH1A3, an enzyme involved in retinoic acid synthesis and the most highly expressed ALDH gene in normal human mammary tissue. In contrast, ALDH1A1 is expressed at low levels in all mammary epithelial cells. These findings raise interesting questions about the reported association of ALDH activity with breast cancer stem cells and breast cancer prognosis.


Subject(s)
Adult Stem Cells/enzymology , Aldehyde Dehydrogenase/metabolism , Mammary Glands, Human/cytology , Adult , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Aldehyde Oxidoreductases , Biomarkers/metabolism , Cells, Cultured , Epithelial Cells/enzymology , Female , Humans , Mammary Glands, Human/enzymology , Retinal Dehydrogenase , Stromal Cells/enzymology , Transcription, Genetic , Young Adult
13.
Br J Nutr ; 110(10): 1743-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23578516

ABSTRACT

The effects of flax meal (FM) on the activity of antioxidant enzymes (superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase (CAT)) in the blood, mammary tissue and ruminal fluid, and oxidative stress indicators (thiobarbituric acid-reactive substances(TBARS) and 1,1-diphenyl-2-picrylhydrazyl-scavenging activity) in the milk, plasma and ruminal fluid of dairy cows were determined.The mRNA abundance of the antioxidant enzymes and oxidative stress-related genes was assessed in mammary tissue. A total of eight Holstein cows were used in a double 4 x 4 Latin square design. There were four treatments in the diet: control with no FM(CON) or 5% FM (5FM), 10% FM (10FM) and 15% FM (15FM). There was an interaction between treatment and time for plasma GPx and CAT activities. Cows supplemented with FM had a linear reduction in TBARS at 2 h after feeding, and there was no treatment effect at 0, 4 and 6 h after feeding. TBARS production decreased in the milk of cows fed the 5FM and 10FM diets. There was a linear increase in nuclear factor (erythroid-derived 2)-like 2 (NFE2L2) mRNA abundance in mammary tissue with FM supplementation.A linear trend for increased mRNA abundance of the CAT gene was observed with higher concentrations of FM. The mRNA abundance of CAT, GPx1, GPx3, SOD1, SOD2, SOD3 and nuclear factor of k light polypeptide gene enhancer in B-cells (NFKB) genes was not affected by the treatment. These findings suggest that FM supplementation can improve the oxidative status of Holstein cows as suggested by decreased TBARS production in ruminal fluid 2 h post-feeding and increased NFE2L2/nuclear factor-E2-related factor 2 (Nrf2) mRNA abundance in mammary tissue.


Subject(s)
Antioxidants/pharmacology , Flax , Mammary Glands, Human/metabolism , Milk/metabolism , Oxidative Stress , Plant Preparations/pharmacology , Rumen/metabolism , Animal Nutritional Physiological Phenomena , Animals , Antioxidants/metabolism , Catalase/blood , Catalase/genetics , Catalase/metabolism , Cattle , Dietary Supplements , Female , Gene Expression/drug effects , Glutathione Peroxidase/blood , Glutathione Peroxidase/metabolism , Humans , Mammary Glands, Human/enzymology , Milk/enzymology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/genetics , RNA, Messenger/metabolism , Seeds , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances
14.
Histopathology ; 60(4): 617-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22260461

ABSTRACT

AIMS: Aldehyde dehydrogenase 1 (ALDH1) in female breast tissue has been linked to stem cells, but little is known about the benign cellular organization in situ. We investigated the distribution of ALDH1-immunoreactive (ALDH1+) cells in histomorphologically benign breast tissue from 28 women with or without breast cancer. METHODS AND RESULTS: ALDH1+ cells were detected in benign tissue of women aged 20-72 years, located most commonly at the luminal and intermediate ductular levels and in the stroma. ALDH1+ cell populations and Ki67+ cell populations were present in separate ductules, both cell types rarely showing epithelial differentiation. ALDH1+ cells were non-reactive to Ki67 and oestrogen receptor. Stromal round/oval ALDH1+ non-leukocyte cells in both age groups expressed contractile protein. There was a lower concentration of luminal and intermediate ductular ALDH1+ cells in postmenopausal women than in premenopausal women, and in cancer patients than in non-cancer patients, and a higher concentration in women receiving exogenous hormones. CONCLUSIONS: This study provides further evidence for the stem cell character of ALDH1+ cells, here in benign breast tissue of cancer and non-cancer patients throughout non-lactating adult life, and contributes evidence of benign stromal ALDH1+ cells. The distribution of ductular ALDH1+ stem cells appears to be influenced by hormonal status.


Subject(s)
Breast Neoplasms/enzymology , Isoenzymes/metabolism , Mammary Glands, Human/enzymology , Retinal Dehydrogenase/metabolism , Stem Cells/enzymology , Adult , Aged , Aldehyde Dehydrogenase 1 Family , Breast Neoplasms/pathology , Female , Humans , Mammary Glands, Human/pathology , Middle Aged
15.
Nutr Cancer ; 64(6): 879-88, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22716309

ABSTRACT

Epidemiological studies have shown that consumption of cabbage and sauerkraut is connected with significant reduction of breast cancer incidences. Estrogens are considered a major breast cancer risk factor and their metabolism by P450 enzymes substantially contributes to carcinogenic activity. The aim of this study was to investigate the effect of cabbage and sauerkraut juices of different origin on the expression profile of the estrogen metabolism key enzymes (CYP1A1, CYP1A2, CYP1B1) in breast cell lines MCF7, MDA-MB-231, and MCF10A. The effects of cabbage juices were compared with that exerted by indole-3-carbinol (I3C) and 3,3'-diindolylmethane (DIM). The treatment with cabbage juices or indoles for 72 h affected the expression of CYP1 family genes in cell-type dependent manner. Their induction was found in all cell lines, but the ratio of CYP1A1 to CYP1B1 was 1.22- to 10.6-fold in favor to CYP1A1 in MCF7 and MCF10A cells. Increased levels of CYP1A2 in comparison with CYP1B1 were also observed in MCF7 cells. In contrast, in MDA-MB-231 cells CYP1B1 was preferentially induced. Since the cell lines investigated differ in invasion capacity, these results support epidemiological observations and partly explain the mechanism of the chemopreventive activity of white cabbage products.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Beverages , Brassica , Breast Neoplasms/enzymology , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Indoles/pharmacology , Mammary Glands, Human/enzymology , Aryl Hydrocarbon Hydroxylases/genetics , Cell Line , Cell Line, Tumor , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1B1 , Female , Fermentation , Gene Expression Regulation, Enzymologic/drug effects , Glucosinolates , Humans , MCF-7 Cells , Mammary Glands, Human/cytology
17.
Am J Physiol Cell Physiol ; 301(5): C1093-103, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21795523

ABSTRACT

Antithrombin, a major anticoagulant, is robustly transported into extravascular compartments where its target proteases are largely unknown. This serpin was previously detected in human milk as complexes with matriptase, a membrane-bound serine protease broadly expressed in epithelial and carcinoma cells, and under tight regulation by hepatocyte growth factor activator inhibitor (HAI)-1, a transmembrane Kunitz-type serine protease inhibitor that forms heat-sensitive complexes with active matriptase. In the current study, we detect, in addition to matriptase-HAI-1 complexes, heat-resistant matriptase complexes generated by nontransformed mammary, prostate, and epidermal epithelial cells that we show to be matriptase-antithrombin complexes. These findings suggest that in addition to HAI-1, interstitial antithrombin participates in the regulation of matriptase activity in epithelial cells. This physiological mechanism appears, however, to largely be lost in cancer cells since matriptase-antithrombin complexes were not detected in all but two of a panel of seven breast, prostate, and ovarian cancer cell lines. Using purified active matriptase, we further characterize the formation of matriptase-antithrombin complex and show that heparin can significantly potentiate the inhibitory potency of antithrombin against matriptase. Second-order rate constants for the inhibition were determined to be 3.9 × 10(3) M(-1)s(-1) in the absence of heparin and 1.2 × 10(5) M(-1)s(-1) in the presence of heparin, a 30-fold increase, consistent with the established role of heparin in activating antithrombin function. Taken together these data suggest that normal epithelial cells employ a dual mechanism involving HAI-1 and antithrombin to control matriptase and that the antithrombin-based mechanism appears lost in the majority of carcinoma cells.


Subject(s)
Antithrombin III/metabolism , Carcinoma/enzymology , Epithelial Cells/enzymology , Proteinase Inhibitory Proteins, Secretory/metabolism , Transcription Factors/antagonists & inhibitors , Antigens, Nuclear/metabolism , Breast Neoplasms/enzymology , Cell Line, Tumor , Epidermis/enzymology , Female , Heparin/pharmacology , Humans , Male , Mammary Glands, Human/enzymology , Ovarian Neoplasms/enzymology , Prostate/enzymology , Prostatic Neoplasms/enzymology , Transcription Factors/metabolism
18.
Am J Pathol ; 176(3): 1241-55, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20110414

ABSTRACT

Recent pregnancy correlates with decreased survival for breast cancer patients compared with non-pregnancy-associated breast cancer. We hypothesize that postpartum mammary involution induces metastasis through wound-healing programs known to promote cancer. It is unknown whether alternatively activated M2 macrophages, immune cells important in wound-healing and experimental tumorigenesis that also predict poor prognosis for breast cancer patients, are recruited to the normal involuting gland. Macrophage markers CD68, CSF-1R, and F4/80 were examined across the pregnancy and involution cycle in rodent and human mammary tissues. Quantitative immunohistochemistry revealed up to an eightfold increase in macrophage number during involution, which returned to nulliparous levels with full regression. The involution macrophages exhibit an M2 phenotype as determined by high arginase-1 and low inducible nitric oxide synthase staining in rodent tissue, and by mannose receptor expression in human breast tissue. M2 cytokines IL-4 and IL-13 also peaked during involution. Extracellular matrix (ECM) isolated from involuting rat mammary glands was chemotactic for macrophages compared with nulliparous mammary ECM. Fibrillar collagen levels and proteolysis increased dramatically during involution, and denatured collagen I acted as a strong chemoattractant for macrophages in cell culture, suggesting proteolyzed fibrillar collagen as a candidate ECM mediator of macrophage recruitment. M2 macrophages, IL-4, IL-13, fibrillar collagen accumulation, and proteolysis of collagen are all components of tumor promotional microenvironments, and thus may mediate promotion of breast cancers arising in the postpartum setting.


Subject(s)
Collagen/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Postpartum Period/immunology , Adult , Aged , Animals , Biomarkers/metabolism , Extracellular Matrix/metabolism , Female , Humans , Macrophages/cytology , Macrophages/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/enzymology , Mammary Glands, Human/cytology , Mammary Glands, Human/enzymology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype , Pregnancy , Rats , Rats, Sprague-Dawley , Species Specificity , Weaning
19.
Stem Cells ; 28(6): 1081-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20506111

ABSTRACT

The major components of the mammary ductal tree are an inner layer of luminal cells, an outer layer of myoepithelial cells, and a basement membrane that separates the ducts from the underlying stroma. Cells in the outer layer express CD10, a zinc-dependent metalloprotease that regulates the growth of the ductal tree during mammary gland development. To define the steps in the human mammary lineage at which CD10 acts, we have developed an in vitro assay for human mammary lineage progression. We show that sorting for CD10 and EpCAM cleanly separates progenitors from differentiated luminal cells and that the CD10-high EpCAM-low population is enriched for early common progenitor and mammosphere-forming cells. We also show that sorting for CD10 enriches sphere-forming cells from other tissue types, suggesting that it may provide a simple tool to identify stem or progenitor populations in tissues for which lineage studies are not currently possible. We demonstrate that the protease activity of CD10 and the adhesion function of beta1-integrin are required to prevent differentiation of mammary progenitors. Taken together, our data suggest that integrin-mediated contact with the basement membrane and cleavage of signaling factors by CD10 are key elements in the niche that maintains the progenitor and stem cell pools in the mammary lineage.


Subject(s)
Mammary Glands, Human/enzymology , Neprilysin/metabolism , Stem Cells/enzymology , Biomarkers , Cell Differentiation , Cell Lineage , Cells, Cultured , Humans , Integrin beta1/metabolism , Mammary Glands, Human/cytology , Stem Cells/cytology
20.
Future Oncol ; 7(6): 753-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21675838

ABSTRACT

Cyclin D1 overexpression is found in more than 50% of human breast cancers and causes mammary cancer in transgenic mice. Dysregulation of cyclin D1 gene expression or function contributes to the loss of normal cell cycle control during tumorigenesis. Recent studies have demonstrated that cyclin D1 conducts additional specific functions to regulate gene expression in the context of local chromatin, promote cellular migration and inhibit mitochondrial metabolism. It is anticipated that these additional functions contribute to the pathology associated with dysregulated cyclin D1 abundance. This article discusses evidence that examines the significance of cyclin D1 in breast cancer with emphasis on its role in breast cancer stem cell expansion.


Subject(s)
Breast Neoplasms/metabolism , Cyclin D1/metabolism , Adult Stem Cells/metabolism , Animals , Cyclin D1/genetics , Cyclin-Dependent Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/enzymology , Protein Isoforms/genetics , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL