Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
Cell Prolif ; 49(4): 460-70, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27323693

ABSTRACT

OBJECTIVES: Frizzled-7 (FZD7) receptor-dependent activation of the canonical Wnt/ß-catenin pathway plays a crucial role in epithelial-to-mesenchymal transition (EMT) and breast cancer metastasis. FZD7 and its co-receptor, low-density lipoprotein receptor-related protein 6 (LRP6), are highly expressed in MDA-MB-231 and T-47D breast cancer cells, and endogenous ligands for FZD7 include Wnt3a and Wnt5a/b. γ-Tocotrienol, a natural isoform of vitamin E, inhibits human breast cancer cell proliferation and EMT. Here, studies have been conducted to investigate the role of the canonical Wnt pathway in mediating inhibitory effects of γ-tocotrienol on EMT in human breast cancer cells. MATERIALS AND METHODS: MDA-MB-231, T-47D and MCF-10A cells were maintained in serum-free defined media containing selected doses of γ-tocotrienol. Cell viability was determined using the MTT colorimetric assay, Western blot analysis was used to measure protein expression and the wound-healing assay was employed to study cell mobility and migration. Immunohistochemical fluorescence staining visualized expression and localization of EMT cell markers. RESULTS: γ-Tocotrienol was found to induce dose-responsive inhibition of MDA-MB-231 and T-47D cell growth at doses that had no effect on immortalized normal MCF-10A mammary epithelial cells. These growth inhibitory effects were associated with suppression in canonical Wnt signalling, reversal of EMT and significant reduction in breast cancer cell motility. CONCLUSIONS: γ-Tocotrienol suppression of metastatic breast cancer cell proliferation and EMT was associated with suppression of the canonical Wnt/ß-catenin signalling pathway.


Subject(s)
Antioxidants/pharmacology , Breast Neoplasms/drug therapy , Breast/drug effects , Chromans/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Vitamin E/analogs & derivatives , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antineoplastic Agents/pharmacology , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Vitamin E/pharmacology , Wnt Proteins/metabolism
2.
Cell Prolif ; 48(4): 421-35, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26096843

ABSTRACT

OBJECTIVES: Aberrant c-Myc activity plays a central role in cancer transformation. γ-tocotrienol is a member of the vitamin E family that displays potent anti-cancer activity. Here, studies were conducted to determine the role of c-Myc in mediating anti-proliferative effects of γ-tocotrienol in mammary cancer cells. MATERIALS AND METHODS: Treatment effects on mouse +SA and human MCF-7 mammary cancer cell proliferation were determined by MTT assay and Ki-67 staining. Protein expression was determined by western blot analysis. Immunofluorescence staining and qRT-PCR were used to characterize cellular c-Myc and MYC levels respectively. RESULTS: Anti-proliferative effects of γ-tocotrienol were associated with reduction in total c-Myc and phosphorylated-c-Myc-serine 62, and increase in phosphorylated-c-Myc-threonine 58 levels. γ-tocotrienol also reduced PI3K/Akt/mTOR and Ras/MEK/Erk mitogenic signalling, cyclin D1 and cyclin-dependent kinase 4 levels, and increased p27 levels. However, γ-tocotrienol had no effect on MYC mRNA levels. γ-tocotrienol also increased levels of FBW7 (E3 ligase that initiates ubiquitination of c-Myc), but had no effect on serine/threonine phosphatase PP2A or isomerase Pin 1 levels. Combined treatment with GSK3α/ß inhibitor LiCl or proteasome inhibitor MG132 blocked γ-tocotrienol-induced reductions in c-Myc. CONCLUSIONS: These findings indicate that anti-proliferative effects of γ-tocotrienol are associated with reduction in c-Myc that results from increase in GSK-3α/ß-dependent ubiquitination and degradation, rather than from reduction in c-Myc synthesis in +SA and MCF-7 mammary cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Chromans/pharmacology , Down-Regulation/drug effects , Proto-Oncogene Proteins c-myc/genetics , Vitamin E/analogs & derivatives , Animals , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Vitamin E/pharmacology , alpha-Tocopherol/administration & dosage , alpha-Tocopherol/pharmacology
3.
Cell Prolif ; 46(5): 538-53, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24033536

ABSTRACT

OBJECTIVES: Dysregulation of Met signalling is associated with malignant transformation. Combined treatment has been shown to reduce Met activation and mammary tumour cell proliferation. Experiments here, were conducted to determine mechanisms involved in mediating anti-cancer effects of combined γ-tocotrienol and SU11274 (Met inhibitor) treatment in various mammary cancer cell lines. MATERIALS AND METHODS: Treatment effects on mouse (+SA) and human (MCF-7, and MDA-MB-231) mammary cancer cell lines, and normal mouse (CL-S1) and human (MCF10A) mammary epithelial cell lines were compared. Cell proliferation and survival were determined by MTT assay and Ki-67 staining; protein expression was determined by western blot analysis. Immunofluorescence staining was also used to characterize expression and localization of multiple epithelial and mesenchymal markers. Cell migration was determined using a wound-healing assay. RESULTS: Combined treatment with γ-tocotrienol and SU11274 resulted in synergistic inhibition of +SA, MCF-7, and MDA-MB-231, but not CL-S1 or MCF10A cell growth that was associated with reduction in Akt STAT1/5 and NFκB activation and corresponding blockade in epithelial-to-mesenchymal transition, as indicated by increased expression of E-cadherin, ß-catenin, and cytokeratins 8/18 (epithelial markers) and corresponding reduction in vimentin (mesenchymal marker) and reduction in cancer cell motility. CONCLUSIONS: Suggest that combined γ-tocotrienol and Met inhibitor treatment may provide benefit in treatment of breast cancers characterized by aberrant Met activity.


Subject(s)
Chromans/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Indoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Sulfonamides/pharmacology , Vitamin E/analogs & derivatives , Animals , Cadherins/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Keratin-18/metabolism , Keratin-8/metabolism , Ki-67 Antigen/chemistry , Ki-67 Antigen/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Vimentin/metabolism , Vitamin E/pharmacology , beta Catenin/metabolism
4.
Cell Prolif ; 44(6): 516-26, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21973114

ABSTRACT

OBJECTIVES: Aberrant Met signalling is associated with aggressive cancer cell phenotypes. γ-tocotrienol displays potent anti-cancer activity that is associated with suppression of HER/ErbB receptor signalling. Experiments were conducted to investigate the effects of γ-tocotrienol treatment on HGF-dependent +SA mammary tumour cell proliferation, upon Met activation. MATERIALS AND METHODS: The +SA cells were maintained in serum-free defined media containing 10 ng/ml HGF as the mitogen. Cell viability was determined using the MTT assay, western blot analysis was used to measure protein expression, and Met expression and activation were determined using immunofluorescent staining. RESULTS AND CONCLUSIONS: Treatment with γ-tocotrienol or Met inhibitor, SU11274, significantly inhibited HGF-dependent +SA cell replication in a dose-responsive manner. Treatment with 4 µmγ-tocotrienol reduced both total Met levels and HGF-induced Met autophosphorylation. In contrast, similar treatment with 5.5 µm SU11274 inhibited HGF-induced Met autophosphorylation, but had no effect on total Met levels. Combined treatment with subeffective doses of γ-tocotrienol (2 µm) and SU11274 (3 µm) resulted in significant inhibition of +SA cell expansion compared to treatment with individual agents alone. These findings show, for the first time, the inhibitory effects of γ-tocotrienol on Met expression and activation, and strongly suggest that γ-tocotrienol treatment may provide significant health benefits in prevention and/or treatment of breast cancer, in women with deregulated HGF/Met signalling.


Subject(s)
Antineoplastic Agents/pharmacology , Chromans/pharmacology , Hepatocyte Growth Factor/antagonists & inhibitors , Mitogens/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Vitamin E/analogs & derivatives , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Hepatocyte Growth Factor/metabolism , Indoles/pharmacology , Mice , Mice, Inbred BALB C , Mitogens/metabolism , Phosphorylation , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-met/metabolism , Sulfonamides/pharmacology , Vitamin E/pharmacology
5.
Cell Prolif ; 43(1): 77-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19922488

ABSTRACT

OBJECTIVES: Previous studies have shown that gamma-tocotrienol induces potent anti-proliferative effects on +SA mammary tumour cells in culture; here, investigations have been conducted to determine its effects on intracellular signalling proteins involved in regulating cell cycle progression. MATERIALS AND METHODS: +SA cells were maintained in mitogen-free defined media containing 0 or 4 micromgamma-tocotrienol, for 48 h to synchronize cell cycle in G(0) phase, and then they were exposed to 100 ng/ml EGF to initiate cell cycle progression. Whole cell lysates were collected at various time points from each treatment group and were prepared for Western blot analysis. RESULTS AND CONCLUSIONS: Treatment with 4 micromgamma-tocotrienol significantly inhibited +SA cell proliferation over a 4-day culture period. Moreover, this treatment resulted in a relatively large reduction in cyclin D1, cyclin dependent kinase (CDK)4, CDK2 and CDK6 levels, between 4 and 24 h after EGF exposure. Tocotrienol treatment also resulted in a relatively large increase in CDK inhibitor (CKI) p27, prior to and after EGF exposure, but had little effect on levels of CKIs, p21 and p15. Tocotrienol treatment also induced a large relative reduction in retinoblastoma (Rb) protein phosphorylation at ser780 and ser807/811. These findings strongly suggest that anti-proliferative effects of gamma-tocotrienol are associated with reduction in cell cycle progression from G(1) to S, as evidenced by increased p27 levels, and a corresponding decrease in cyclin D1, CDK2, CDK4, CDK6 and phosphorylated Rb levels.


Subject(s)
Cell Cycle/drug effects , Chromans/pharmacology , Mammary Neoplasms, Experimental/metabolism , Vitamin E/analogs & derivatives , Animals , Cell Line , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epidermal Growth Factor/pharmacology , Female , Mammary Glands, Animal/metabolism , Mice , Phosphorylation , Resting Phase, Cell Cycle , Vitamin E/pharmacology
6.
Cell Prolif ; 39(6): 563-74, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17109639

ABSTRACT

The antiproliferative effects of gamma-tocotrienol are associated with suppression in epidermal growth factor (EGF)-dependent phosphatidylinositol-3-kinase (PI3K)/PI3K-dependent kinase-1 (PDK-1)/Akt mitogenic signalling in neoplastic mammary epithelial cells. Studies were conducted to investigate the direct effects of gamma-tocotrienol treatment on specific components within the PI3K/PDK-1/Akt mitogenic pathway. +SA cells were grown in culture and maintained in serum-free media containing 10 ng/ml EGF as a mitogen. Treatment with 0-8 microm gamma-tocotrienol resulted in a dose-responsive decrease in the +SA cell growth and a corresponding decrease in phospho-Akt (active) levels. However, gamma-tocotrienol treatment had no direct inhibitory effect on Akt or PI3K enzymatic activity, suggesting that the inhibitory effects of gamma-tocotrienol occur upstream of PI3K, possibly at the level of the EGF-receptor (ErbB1). Additional studies were conducted to determine the effects of gamma-tocotrienol on ErbB receptor activation. Results showed that gamma-tocotrienol treatment had little or no effect on ErbB1 or ErbB2 receptor tyrosine phosphorylation, a prerequisite for substrate interaction and signal transduction, but did cause a significant and progressive decrease in the ErbB3 tyrosine phosphorylation. Because ErbB1 or ErbB2 receptors form heterodimers with the ErbB3 receptor, and ErbB3 heterodimers have been shown to be the most potent activators of PI3K, these findings strongly suggest that the antiproliferative effects of gamma-tocotrienol in neoplastic +SA mouse mammary epithelial cells are mediated by a suppression in ErbB3-receptor tyrosine phosphorylation and subsequent reduction in PI3K/PDK-1/Akt mitogenic signalling.


Subject(s)
Adenocarcinoma/metabolism , Chromans/pharmacology , Mammary Neoplasms, Animal/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Vitamin E/analogs & derivatives , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Epithelial Cells/cytology , Female , Mice , Mice, Inbred BALB C , Mitosis/drug effects , Mitosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Vitamin E/pharmacology , Vitamins/pharmacology , alpha-Tocopherol/pharmacology
7.
Cell Prolif ; 38(3): 153-63, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15985060

ABSTRACT

Electromagnetic ultra-wideband pulses (UWB) or nanopulses, are generated by a wide range of electronic devices used in communications and radar technology. However, the specific effects of nanopulse exposure on cell growth and function have not been extensively investigated. Here, studies have been conducted to determine the effects of prolonged exposure to non-ionizing, low to moderate intensity nanopulses on the growth of pre-neoplastic CL-S1 mammary epithelial cells in vitro. Cells were grown in culture and maintained in serum-free defined medium containing 10 ng/ml EGF and 10 microg/ml insulin as comitogens. Studies showed that 0.25-3.0 h exposure to nanopulses of 18 kV/m field intensity, 1 kHz repetition rate and 10 ns pulse width had no effect on CL-S1 cell growth or viability during the subsequent 72-h culture period. However, exposure to similar nanopulses for prolonged periods of time (4-6 h) resulted in a significant increase in cell proliferation, as compared to untreated controls. Additional studies showed that nanopulse exposure enhanced CL-S1 cell growth when cells were maintained in media containing only EGF, but had no effect on cells maintained in defined media that were mitogen-free or containing only insulin. Studies also showed that the growth-promoting effects of nanopulse exposure were associated with a relatively large increase in intracellular levels of phospho-MEK1 (active) and phospho-ERK1/2 (active) in these cells. These findings demonstrate that prolonged exposure to moderate levels of UWB enhanced EGF-dependent mitogenesis, and that this growth-promoting effect appears to be mediated by enhanced activation of the mitogen-activated protein kinase (MAPK) signalling pathway in pre-neoplastic CL-S1 mammary epithelial cells.


Subject(s)
Cell Division/radiation effects , Epithelial Cells/radiation effects , Mammary Neoplasms, Animal/pathology , Precancerous Conditions/pathology , Radiation , Animals , Cell Count , Cell Line, Tumor , Dose-Response Relationship, Radiation , Epithelial Cells/cytology , MAP Kinase Signaling System/radiation effects , Mice , Mice, Inbred BALB C
8.
Cell Prolif ; 34(6): 347-57, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11736999

ABSTRACT

Tocopherols and tocotrienols represent the two subclasses within the vitamin E family of compounds. However, tocotrienols are significantly more potent than tocopherols in suppressing epidermal growth factor (EGF)-dependent normal mammary epithelial cell growth. EGF is a potent mitogen for normal mammary epithelial cells and an initial event in EGF-receptor mitogenic-signalling is protein kinase C (PKC) activation. Studies were conducted to determine if the antiproliferative effects of specific tocopherol and tocotrienol isoforms are associated with a reduction in EGF-receptor mitogenic signalling and/or PKC activation. Normal mammary epithelial cells isolated from midpregnant BALB/c mice were grown in primary culture, and maintained on serum-free media containing 10 ng/mL EGF as a mitogen, and treated with various doses (0-250 microm) of alpha-, gamma-, or delta-tocopherol or alpha-, gamma-, or delta-tocotrienol. Treatment with growth inhibitory doses of delta-tocopherol (100 microm), alpha-tocotrienol (50 microm), or gamma- or delta-tocotrienol (10 microm) did not affect EGF-receptor levels, EGF-induced EGF-receptor tyrosine kinase activity, or total intracellular levels of PKC(alpha). However, these treatments were found to inhibit EGF-induced PKC(alpha) activation as determined by its translocation from the cytosolic to membrane fraction. Treatment with 250 microm alpha- or gamma-tocopherol had no affect on EGF-receptor mitogenic signalling or cell growth. These findings demonstrate that the inhibitory effects of specific tocopherol and tocotrienol isoforms on EGF-dependent normal mammary epithelial cell mitogenesis occurs downstream from the EGF-receptor and appears to be mediated, at least in part, by a reduction in PKC(alpha) activation.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/enzymology , Protein Kinase C/metabolism , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Blotting, Western , Cell Division/drug effects , Cells, Cultured , Chromans/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Female , Mice , Mice, Inbred BALB C , Pregnancy , Time Factors , Tocotrienols , alpha-Tocopherol/pharmacology , gamma-Tocopherol/pharmacology
9.
Neurosci Lett ; 306(3): 141-4, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11406315

ABSTRACT

Mu opioid receptors occur throughout the brain, but central sites where ligand neuromodulatory effects occur during glucopenia have not been identified. The present studies investigated whether septal, preoptic, and hypothalamic neurons that express immunoreactivity for this receptor are transcriptionally activated in response to the glucose antimetabolite, 2-deoxy-D-glucose (2DG), and if intracerebroventricular (icv) administration of the selective mu receptor antagonist, CTOP, modifies this functional response to glucose substrate imbalance. Neurons labeled for mu receptor-immunoreactivity (-ir) were observed in the lateral septal nucleus (LS), medial septum (MS), anterior division of the stria terminalis (BSTa), median preoptic nucleus (MEPO), medial preoptic nucleus (MPN), parastrial nucleus (PS), anterior hypothalamic periventricular nucleus (PVa), and lateral hypothalamic area (LPO). 2DG injection (400 mg/kg i.p.) resulted in co-labeling of mu receptor-positive neurons in the LS, MS, BSTa, MEPO, PVa, and LPO for nuclear Fos-ir. Icv delivery of CTOP decreased mean numbers of co-labeled neurons in the LS, MS, BSTa, and MEPO. These results provide evidence for transactivational effects of glucopenia on mu opioid receptor-expressing neurons within the septum, preoptic area, and hypothalamus, and suggest that the functional status of these receptors within discrete septopreoptic sites may be critical for maximal glucoprivic induction of the Fos stimulus-transcription cascade within local cells. These results thus support the view that the neural loci described above may serve as substrates for regulatory effects of mu opioid receptor ligands on central compensatory activities during acute glucose deprivation.


Subject(s)
Glucose/deficiency , Hypothalamic Area, Lateral/chemistry , Preoptic Area/chemistry , Proto-Oncogene Proteins c-fos/analysis , Receptors, Opioid, mu/analysis , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Animals , Antibodies , Antimetabolites/pharmacology , Deoxyglucose/pharmacology , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Male , Preoptic Area/drug effects , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/immunology , Rats , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/immunology , Septal Nuclei/chemistry , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Septum of Brain/chemistry , Septum of Brain/drug effects , Septum of Brain/metabolism
10.
Neuroendocrinology ; 73(6): 369-77, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11408778

ABSTRACT

Hyperphagic and hypothalamic neuroendocrine responses to acute glucose deprivation are modified by the ovarian steroid estradiol (E). Observations of genomic activation of catecholaminergic (CA) neurons in the hindbrain lateral reticular nucleus, nucleus of the solitary tract, and area postrema (AP) by glucopenia support their potential function in pathways mediating regulatory effects of this metabolic challenge within the brain. Expression of E receptors by these cells suggests that their activity may be sensitive to steroid modulation during glucopenia. The present studies investigated the role of E on transcriptional activation of caudal brainstem CA neurons by the glucose antimetabolite, 2-deoxy-D-glucose (2DG). Ovariectomized rats were implanted with s.c. Silastic capsules containing E (30 or 250 microg/ml) or sesame oil, and injected i.p. 7 days later with 400 mg 2DG/kg or saline. Tyrosine hydroxylase (TH)-immunoreactive (-ir) neurons in the C(1)/A1, C2, C3, A2, A5, and A6 cell groups and AP were colabeled for Fos following antimetabolite administration, whereas vehicle injection resulted in negligible nuclear staining of these cells. With the exception of A2, A6, and AP cells, mean numbers of Fos- and TH-/Fos-ir-positive neurons in these brain sites did not differ between E- and sesame oil-implanted groups. Numbers of TH-positive A2 and A6 neurons that expressed Fos in response to 2DG were significantly greater in rats implanted with the high E dose vs. either the low steroid dose or sesame oil. These results show that the magnitude of cellular Fos labeling within discrete hindbrain CA neuron populations varies in accordance with circulating E levels. These findings suggest that E may exert potential modulatory effects on glucoprivic activation of the Fos stimulus/transcription cascade and consequent compensatory genomic responses within specific areas of the female rat caudal brainstem.


Subject(s)
Brain Stem/cytology , Catecholamines/biosynthesis , Estradiol/pharmacology , Glucose/deficiency , Neurons/physiology , Animals , Deoxyglucose/pharmacology , Drug Implants , Estradiol/administration & dosage , Female , Neurons/chemistry , Ovariectomy , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tyrosine 3-Monooxygenase/analysis
11.
Neuroreport ; 12(3): 531-4, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11234758

ABSTRACT

Reports that glucose antimetabolite treatment elicits hyperphagia and hyperglycemia suggest that decreased oxidation of this energy substrate elicits compensatory responses that enhance cellular fuel availability. Neurons the lateral hypothalamic area (LHA) synthesize the orectic neuropeptide, orexin-A (ORX-A). The present study evaluated the functional responsiveness of orexinergic neurons to glucopenia by investigating whether these cells express the genomic regulatory protein, Fos, in response to glucoprivation. Adult male rats were sacrificed 2h after i.p. (400 mg/kg) or intracerebroventricular (i.c.v.; 100 microg) administration of the antimetabolite, 2-deoxy-D-glucose (2DG) or saline. Sections through the LHA, from the level of the paraventricular nucleus (PVN) to the posterior hypothalamic area (PHA), were processed by dual-label immunocytochemistry for Fos- and OXY-A-immunoreactivity (-ir). Although orexinergic neurons expressed negligible Fos-ir following vehicle administration, dual-labeled ORX-A neurons were observed in the LHA, as well as the dorsomedial hypothalamic nucleus (DMN) and PHA, in both drug-treated groups. Bilateral cell counts from representative levels of the LHA, DMN, and PHA showed that in each structure, a greater proportion of ORX-A neurons were immunostained for Fos in response to systemic than following i.c.v. treatment with 2DG. These results provide evidence for the transcriptional activation of hypothalamic ORX-A neurons by diminished glucose availability, data that suggest that these cells may function within central pathways that govern adaptive responses to deficits of this substrate fuel. The findings also support the view that a proportion of this phenotypic population is responsive to glucoprivic stimuli of central origin.


Subject(s)
Carrier Proteins/analysis , Glucose/metabolism , Hypothalamus/cytology , Intracellular Signaling Peptides and Proteins , Neurons/metabolism , Neuropeptides/analysis , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Antibodies , Antimetabolites/pharmacokinetics , Carrier Proteins/immunology , Deoxyglucose/pharmacokinetics , Dorsomedial Hypothalamic Nucleus/cytology , Energy Metabolism/physiology , Homeostasis/drug effects , Homeostasis/physiology , Hypothalamic Area, Lateral/cytology , Hypothalamus, Posterior/cytology , Male , Neurons/chemistry , Neuropeptides/immunology , Orexins , Oxidation-Reduction , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley
12.
Proc Soc Exp Biol Med ; 224(4): 292-301, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964265

ABSTRACT

Studies were conducted to determine the comparative effects of tocopherols and tocotrienols on preneoplastic (CL-S1), neoplastic (-SA), and highly malignant (+SA) mouse mammary epithelial cell growth and viability in vitro. Over a 5-day culture period, treatment with 0-120 microM alpha- and gamma-tocopherol had no effect on cell proliferation, whereas growth was inhibited 50% (IC50) as compared with controls by treatment with the following: 13, 7, and 6 microM tocotrienol-rich-fraction of palm oil (TRF); 55, 47, and 23 microM delta-tocopherol; 12, 7, and 5 microM alpha-tocotrienol; 8, 5, and 4 microM gamma-tocotrienol; or 7, 4, and 3 microM delta-tocotrienol in CL-S1, -SA and +SA cells, respectively. Acute 24-hr exposure to 0-250 microM alpha- or gamma-tocopherol (CL-S1, -SA, and +SA) or 0-250 microM delta-tocopherol (CL-S1) had no effect on cell viability, whereas cell viability was reduced 50% (LD50) as compared with controls by treatment with 166 or 125 microM delta-tocopherol in -SA and +SA cells, respectively. Additional LD50 doses were determined as the following: 50, 43, and 38 microM TRF; 27, 28, and 23 microM alpha-tocotrienol; 19, 17, and 14 microM gamma-tocotrienol; or 16, 15, or 12 microM delta-tocotrienol in CL-S1, -SA, and +SA cells, respectively. Treatment-induced cell death resulted from activation of apoptosis, as indicated by DNA fragmentation. Results also showed that CL-S1, -SA, and +SA cells preferentially accumulate tocotrienols as compared with tocopherols, and this may partially explain why tocotrienols display greater biopotency than tocopherols. These data also showed that highly malignant +SA cells were the most sensitive, whereas the preneoplastic CL-S1 cells were the least sensitive to the antiproliferative and apoptotic effects of tocotrienols, and suggest that tocotrienols may have potential health benefits in preventing and/or reducing the risk of breast cancer in women.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Mammary Neoplasms, Animal/pathology , Precancerous Conditions/pathology , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/pathology , Mammary Neoplasms, Animal/drug therapy , Mice , Neoplasm Invasiveness/pathology , Precancerous Conditions/drug therapy , Time Factors , Tumor Cells, Cultured , Vitamin E/chemistry , Vitamin E/therapeutic use
13.
Lipids ; 35(2): 171-80, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10757548

ABSTRACT

Studies were conducted to determine the comparative effects of tocopherols and tocotrienols on normal mammary epithelial cell growth and viability. Cells isolated from midpregnant BALB/c mice were grown within collagen gels and maintained on serum-free media. Treatment with 0-120 microM alpha- and gamma-tocopherol had no effect, whereas 12.5-100m microM tocotrienol-rich fraction of palm oil (TRF), 100-120 microM delta-tocopherol, 50-60 microM alpha-tocotrienol, and 8-14 microM gamma- or delta-tocotrienol significantly inhibited cell growth in a dose-responsive manner. In acute studies, 24-h exposure to 0-250 microM alpha-, gamma-, and delta-tocopherol had no effect, whereas similar treatment with 100-250 microM TRF, 140-250 microM alpha-, 25-100 microM gamma- or delta-tocotrienol significantly reduced cell viability. Growth-inhibitory doses of TRF, delta-tocopherol, and alpha-, gamma-, and delta-tocotrienol were shown to induce apoptosis in these cells, as indicated by DNA fragmentation. Results also showed that mammary epithelial cells more easily or preferentially took up tocotrienols as compared to tocopherols, suggesting that at least part of the reason tocotrienols display greater biopotency than tocopherols is because of greater cellular accumulation. In summary, these findings suggest that the highly biopotent gamma- and delta-tocotrienol isoforms may play a physiological role in modulating normal mammary gland growth, function, and remodeling.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Mammary Glands, Animal/drug effects , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Chromans/pharmacology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Inhibitory Concentration 50 , Isomerism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , Palm Oil , Plant Oils/pharmacology , Tocotrienols
14.
Cancer Chemother Pharmacol ; 44(2): 152-8, 1999.
Article in English | MEDLINE | ID: mdl-10412950

ABSTRACT

PURPOSE: P-glycoprotein is a 170-kDa plasma membrane multidrug transporter that actively exports cytotoxic substances from cells. Overexpression of P-glycoprotein by tumor cells is associated with a multidrug-resistant phenotype. Immunosuppressive agents such as cyclosporins and macrolides, have been shown to attenuate P-glycoprotein activity. However, the mechanism by which some immunosuppressants inhibit P-glycoprotein function has not been determined. Since cyclosporin and macrolide immunosuppressants inhibit calcineurin (CaN) phosphatase and FKBP12 peptideprolyl isomerase (FKBP12 PPI) activity, studies were conducted to determine if these effects are directly related to the inhibitory effects these immunosuppressants have on P-glycoprotein function. METHODS: Western blot analysis was performed to assess CaN and FKBP12 protein levels in P-glycoprotein-negative (MCF-7) and -positive (MCF-7/Adr) breast cancer cell lines. P-glycoprotein function was determined by intracellular doxorubicin accumulation and/or cytotoxicity assays before and after CaN and FKBP12 were independently inhibited by pharmacological antagonists. RESULTS: CaN and FKBP12 levels were similar in MCF-7 and MCF-7/Adr cells. P-glycoprotein function was not affected by treatment of P-glycoprotein-expressing MCF-7/Adr cells with CaN and FKBP12 antagonists. CONCLUSIONS: These results demonstrate that the inhibitory effects of immunosuppressive agents on P-glycoprotein function are independent of CaN or FKBP12 PPI activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Calcineurin Inhibitors , Immunophilins/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Peptidylprolyl Isomerase/antagonists & inhibitors , Phosphoric Monoester Hydrolases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Tacrolimus Binding Proteins , Tumor Cells, Cultured
15.
Neuroendocrinology ; 69(3): 181-90, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10087450

ABSTRACT

Neuronal nitric oxide synthase, e.g. NADPH diaphorase (NADPH-d), catalyzes formation of the free radical, nitric oxide (NO), and occurs within brain structures that have functional significance for energy fuel homeostasis. The following studies examined whether populations of NADPH-d-positive neurons in the hypothalamus and nearby preoptic area express immunoreactivity for the nuclear transcription factor, Fos, in response to glucose substrate imbalance. Eight days after bilateral ovariectomy (OVX) and subcutaneous implantation of silastic capsules containing 30 microgram estradiol benzoate/ml, female rats were injected i.p. with the glucose antimetabolite, 2-deoxy-D-glucose (2DG; 400 mg/kg), or the vehicle, saline. The animals were sacrificed by transcardial perfusion 2 h after these treatments. Sections at 150-micrometer intervals throughout preoptic area and anterior and tuberal regions of the hypothalamus were processed for dual cytoplasmic NADPH-d enzyme activity and nuclear Fos-immunoreactivity (-ir). The glucose antimetabolite elicited expression of nuclear Fos-ir by NADPH-d-positive neurons in several neural structures, including the medial preoptic area, median preoptic nucleus, anterior commissural, periventricular magnocellular supraoptic nucleus, paraventricular nucleus, and medial part of the bed nucleus of the stria terminalis. In contrast, the extensive populations of NADPH-d-positive neurons in the ventromedial hypothalamic nucleus and lateral hypothalamic area showed very little immunolabeling for Fos in response to glucoprivation. This demonstration of nuclear immunoreactivity for Fos suggests that cellular glucopenia elicits the transcriptional activation, via AP-1 regulatory sites, of multiple populations of hypothalamic neurons characterized by the functional capacity to generate NO, and thus that this gaseous neurotransmitter may fulfill a role(s) in central neural mechanisms governing regulation of compensatory motor responses to metabolic imbalance.


Subject(s)
Glucose/administration & dosage , Hypothalamus/chemistry , NADPH Dehydrogenase/analysis , Neurons/chemistry , Preoptic Area/chemistry , Proto-Oncogene Proteins c-fos/analysis , Animals , Cell Nucleus/chemistry , Cytoplasm/enzymology , Deoxyglucose/pharmacology , Drug Implants , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Female , Histocytochemistry , Hypothalamus/enzymology , Immunohistochemistry , Neurons/drug effects , Neurons/enzymology , Ovariectomy , Preoptic Area/enzymology , Rats , Rats, Sprague-Dawley
16.
J Toxicol Environ Health A ; 55(2): 93-106, 1998 Sep 25.
Article in English | MEDLINE | ID: mdl-9761130

ABSTRACT

Concerns have been raised over the use of iodine for disinfecting drinking water on extended space flights. Most fears revolve around effects of iodide on thyroid function. iodine (I2) is the form used in drinking-water disinfection. Risk assessments have treated the various forms of iodine as if they were toxicologically equivalent. Recent experiments conducted in rats found that administration of iodine as I- (iodide) versus I2 had opposite effects on plasma thyroid hormone levels. I2-treated animals displayed elevated thyroxine (T4) and thyroxine/triiodothyronine (T/T3) ratios, whereas those treated with I- displayed no change or reduced plasma concentrations of T4 at concentrations in drinking water of 30 or 100 mg/L. The study herein was designed to assess whether similar effects would be seen in humans as were observed in rats. A 14-d repeated-dose study utilizing total doses of iodine in the two forms at either 0.3 or 1 mg/kg body weight was conducted with 33 male volunteers. Thyroid hormones evaluated included T4, T3, and thyroid-stimulating hormone (TSH). TSH was significantly increased by the high dose of both I2 and I-, as compared to the control. Decreases in T4 were observed with dose schedules with I- and I2, but none were statistically significant compared to each other, or compared to the control. This human experiment failed to confirm the differential effect of I2 on maintenance of serum T4 concentrations relative to the effect of I- that was observed in prior experiments in rats. However, based on the elevations in TSH, there should be some concern over the potential impacts of chronic consumption of iodine in drinking water.


Subject(s)
Disinfectants/pharmacology , Iodides/pharmacology , Iodine/pharmacology , Thyroid Gland/drug effects , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Drinking , Fresh Water , Humans , Iodides/blood , Iodides/urine , Iodine/blood , Iodine/urine , Male , Risk Assessment , Thyroid Gland/physiology , Water Purification
17.
J Neuroendocrinol ; 10(10): 769-76, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9792328

ABSTRACT

In female mammals, the preovulatory luteinizing hormone (LH) 'surge' elicits ovulation and the subsequent transformation of Graafian follicles into corpora lutea, and is thus a critical component of successful reproduction. In light of evidence that this surge is impaired as a consequence of caloric restriction, the following experiments utilized pharmacological strategies to determine whether glucose substrate homeostasis influences the magnitude and/or duration of this pivotal hormonal event. Groups of oestrogen-and progesterone-primed ovariectomized (OVX) rats were injected intravenously (i.v.) with the glucose antimetabolite, 2-deoxy-D-glucose (2DG: 100 or 400 mg/kg), or the vehicle, saline, prior to onset of the expected LH surge. Other rats were pretreated with 2DG (100 microg/rat) or saline by an intracerebroventricular (i.c.v) route. While glucoprivation did not abolish the afternoon LH surge in these animals, mean plasma LH levels were significantly decreased in groups injected with the higher i.v. dose of 2DG or treated with this drug by an i.c.v route, relative to their vehicle-injected controls. In other studies, i.c.v delivery of the opioid receptor antagonist, naltrexone (NALT), partially reversed the inhibitory effects of 2DG on the gonadal steroid-induced LH surge. Dual-label immunocytochemistry of tissue sections from the preoptic area and anterior hypothalamus of OVX, steroid-primed rats revealed nuclear Fos-immunoreactivity (-ir) in a subpopulation of gonadotropin-releasing hormone-(GnRH-)immunopositive neurones prior to maximal preovulatory LH release. Animals pretreated with 2DG i.c.v showed a significant decrease in mean numbers of GnRH neurones exhibiting Fos-ir, whereas coadministration of 2DG and NALT resulted in numbers of double-labelled neurones that were similar to those detected in the non-drug-treated controls. These studies show that magnitude of the LH surge is decreased by glucose substrate imbalance, and that regulatory effects of this metabolic challenge on the reproductive neuroendocrine axis is correlated with alterations in the transcriptional activation of preoptic GnRH neurones by gonadal steroid positive feedback. The present results also support a role for central opiatergic neurotransmission in glucoprivic regulation of cyclic LH secretion in this animal model.


Subject(s)
Endorphins/physiology , Genes, fos/physiology , Glucose/physiology , Gonadotropin-Releasing Hormone/physiology , Luteinizing Hormone/biosynthesis , Neurons/physiology , Preoptic Area/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Dose-Response Relationship, Drug , Estrogens/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry , Neurons/drug effects , Ovariectomy , Preoptic Area/cytology , Radioimmunoassay , Rats , Rats, Sprague-Dawley
18.
Cell Prolif ; 31(1): 35-46, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9666817

ABSTRACT

Epidermal growth factor (EGF) is a potent mitogen for normal mouse mammary epithelial cells grown in primary culture. EGF activation of the EGF-receptor (EGF-R) induces intrinsic tyrosine kinase activity which results in EGF-R autophosphorylation and tyrosine phosphorylation of other intracellular substrates involved in EGF-R signal transduction. Genistein and erbstatin are anticancer agents which have been shown to be potent tyrosine kinase inhibitors. However, the effects of these compounds in modulating EGF-dependent normal mammary epithelial cell proliferation is presently unknown. Therefore, studies were conducted to determine the effects of genistein and erbstatin on EGF-dependent proliferation, and EGF-R levels and autophosphorylation in normal mouse mammary epithelial cells grown in primary culture and maintained in serum-free media. Chronic treatment with 6.25-100 microM genistein or 1-16 microM erbstatin significantly decreased EGF-dependent mammary epithelial cell proliferation in a dose-responsive manner. However, the highest doses of genistein (100 microM) and erbstatin (16 microM) were found to be cytotoxic. Additional studies showed that acute treatment with 6.25-400 microM genistein did not affect EGF-R levels or EGF-induced EGF-R autophosphorylation, while acute treatment with 1-64 microM erbstatin caused a slight reduction in EGF-R levels, but had no effect on EGF-dependent EGF-R autophosphorylation in these cells. In contrast, chronic treatment with similar doses of genistein or erbstatin resulted in a large dose-responsive decrease in EGF-R levels, and a corresponding decrease in total cellular EGF-R autophosphorylation intensity. These results demonstrate that the inhibitory effects of chronic genistein and erbstatin treatment on EGF-dependent mammary epithelial cell proliferation is not due to a direct inhibition of EGF-R tyrosine kinase activity, but results primarily from a down-regulation in EGF-R levels and subsequent decrease in mammary epithelial cell mitogenic-responsiveness to EGF stimulation.


Subject(s)
Down-Regulation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Genistein/pharmacology , Growth Inhibitors/pharmacology , Hydroquinones/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Female , Mammary Glands, Animal , Mice , Mice, Inbred BALB C
19.
Proc Soc Exp Biol Med ; 217(2): 180-7, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9452142

ABSTRACT

Epidermal growth factor (EGF)- and insulin-dependent mammary epithelial cell mitogenesis is mediated by specific tyrosine kinase receptors. Receptor tyrosine kinase activity is highly regulated in normal cells, whereas amplification of intracellular protein tyrosine phosphorylation is associated with abnormal growth and/or neoplastic transformation. Since protein tyrosine phosphatases (PTPs) are involved in regulating receptor tyrosine kinase signaling, studies were conducted to determine the effects of the PTP inhibitors, vanadate and pervanadate, on mitogen-receptor signal transduction and cell growth. Mammary epithelial cells isolated from midpregnant BALB/c mice were grown within collagen gels and maintained on serum-free media. Treatment with 2-8 microM vanadate or pervanadate greatly increased intracellular protein tyrosine phosphorylation. However, in the presence of optimal mitogenic stimulation (10 ng/ml EGF and 10 microg/ml insulin), these treatments induced a slight, but significant decrease in cell growth. In contrast, these treatments significantly increased mammary epithelial cell growth, albeit less than optimally, under submitogenic culture conditions (500 pg/ml EGF and 10 microg/ml insulin). Neither vanadate nor pervanadate was found to mimic the mitogenic actions of EGF and/or insulin in these cells. The growth-stimulatory effects of PTP inhibitors in submitogenic conditions appear to result from enhanced receptor tyrosine kinase mitogenic signaling, whereas PTP inhibitor attenuation of optimal cell growth may be due to the suppression of PTP activity associated with cell cycle progression. In addition, treatment with PTP inhibitors was not found to stimulate anchorage-independent growth, as determined by the inability of single cells to form colonies in soft agarose. In conclusion, these data demonstrate that optimal mitogen-dependent mammary epithelial cell growth requires both receptor tyrosine kinase and PTP activity. Furthermore, PTP inhibitor-induced amplification of receptor tyrosine kinase mitogenic signaling is not in itself sufficient to induce enhanced cell growth or phenotypic expression of neoplastic transformation.


Subject(s)
Epidermal Growth Factor/pharmacology , Insulin/pharmacology , Mammary Glands, Animal/cytology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Cell Division/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Female , Mice , Mice, Inbred BALB C , Phosphotyrosine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Vanadates/pharmacology
20.
Proc Soc Exp Biol Med ; 213(1): 65-70, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8820825

ABSTRACT

Mammary epithelial cells isolated from midpregnant BALB/c mice were grown within collagen gels and maintained in serum-free media containing 10 ng/ml epidermal growth factor (EGF) for an 8-day culture period. Western blot and scanning densitometric image analysis showed the presence of protein kinase C(PKC)alpha (82 kDa), delta(75 kDa), eta(90 and 78 kDa), and zeta(82, 74, and 65 kDa), whereas PKCbeta, gamma, and theta were not detected in either the cytosolic or membrane fractions in these cells. Cytosolic and membrane levels of PKCalpha and 82 kDa PKCzeta band progressively increased throughout the 8-day culture period. During this same time, cytosolic PKCdelta levels decreased, while membrane levels of PKCdelta showed no change. Cytosolic and membrane levels of PKCeta and the 74- and 65-kDa PKCzeta bands displayed some fluctuations but remained relatively constant during the 8-day culture period. Other studies showed that 24-hr treatment with 100 nM of phorbol 12-myristate 13-acetate (PMA), resulted in the downregulation of PKCalpha, delta, and eta, and the 82-kDa PKCzeta band. However, PMA treatment had no effect on cytosolic and membrane levels of the 74- and 65-kDa PKCzeta bands. Since PKC activation is associated with hormone- and growth factor-dependent mammary epithelial cell proliferation, these findings suggest that increases and/or decreases in the relative levels of the different PKC isoenzymes in proliferating cells may indicate their possible role in mediating or regulating EGF-dependent mitogenesis.


Subject(s)
Isoenzymes/biosynthesis , Mammary Glands, Animal/enzymology , Protein Kinase C/biosynthesis , Animals , Autoradiography/methods , Blotting, Western/methods , Cell Membrane/enzymology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cytosol/enzymology , Epithelium/drug effects , Epithelium/enzymology , Female , Isoenzymes/drug effects , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mice , Mice, Inbred BALB C , Phorbol Esters/pharmacology , Pregnancy , Protein Kinase C/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...