Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Curr Pharm Teach Learn ; 11(7): 723-728, 2019 07.
Article in English | MEDLINE | ID: mdl-31227096

ABSTRACT

BACKGROUND AND PURPOSE: In addition to clinical knowledge, teamwork and critical thinking are skills necessary to be successful during advanced pharmacy practice experiences (APPEs). One way that educators can help students to achieve these skills is with the utilization of educational games. EDUCATIONAL ACTIVITY AND SETTING: Faculty from different departments worked together to develop an educational activity modeled after the escape room game concept for third year pharmacy students enrolled in a pre-APPE readiness course. FINDINGS: The knowledge pre-assessment mean was 81 ±â€¯11.6, with a range of 53 to 105. The mean score following the escape game activity was 79 ±â€¯14.5, with a range of 41 to 100. On average, students scored 3 points lower on the post-assessment (-2.8 ±â€¯13.4). Despite the decrease in overall mean scores from pre-assessment to post-assessment, the overwhelming majority of students (96%, n = 51) felt that this exercise improved clinical skills and facilitated learning. SUMMARY: The escape room activity was developed in such a way that it focused on teamwork, critical thinking, problem solving, and the integration of didactic coursework with the intent that the students could apply their knowledge in a simulated scenario. The students viewed the activity in an overwhelmingly positive light, and their perceptions of the impact of the activity on their ability to think critically and integrate content from their previous courses indicated that the game format has the potential to impact student skills in these areas.


Subject(s)
Clinical Competence/standards , Games, Experimental , Licensure, Pharmacy , Students, Pharmacy/psychology , Clinical Competence/statistics & numerical data , Education, Pharmacy/methods , Education, Pharmacy/statistics & numerical data , Humans , Students, Pharmacy/statistics & numerical data
2.
Cancer Res ; 73(14): 4406-17, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23674500

ABSTRACT

Elevated oxidative stress is observed more frequently in cancer cells than in normal cells. It is therefore expected that additional exposure to a low level of reactive oxygen species (ROS) will push cancer cells toward death, whereas normal cells might maintain redox homeostasis through adaptive antioxidant responses. We previously showed that parthenolide enhances ROS production in prostate cancer cells through activation of NADPH oxidase. The present study identifies KEAP1 as the downstream redox target that contributes to parthenolide's radiosensitization effect in prostate cancer cells. In vivo, parthenolide increases radiosensitivity of mouse xenograft tumors but protects normal prostate and bladder tissues against radiation-induced injury. Mechanistically, parthenolide increases the level of cellular ROS and causes oxidation of thioredoxin (TrX) in prostate cancer cells, leading to a TrX-dependent increase in a reduced state of KEAP1, which in turn leads to KEAP1-mediated PGAM5 and Bcl-xL (BCL2L1) degradation. In contrast, parthenolide increases oxidation of KEAP1 in normal prostate epithelial cells, leading to increased Nrf2 (NFE2L2) levels and subsequent Nrf2-dependent expression of antioxidant enzymes. These results reveal a novel redox-mediated modification of KEAP1 in controlling the differential effect of parthenolide on tumor and normal cell radiosensitivity. Furthermore, they show it is possible to develop a tumor-specific radiosensitizing agent with radioprotective properties in normal cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Radiation-Sensitizing Agents/pharmacology , Sesquiterpenes/pharmacology , Animals , Antioxidants/metabolism , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Kelch-Like ECH-Associated Protein 1 , Male , Mice , Mice, Nude , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Phosphoprotein Phosphatases , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Random Allocation , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Ubiquitin/metabolism , Xenograft Model Antitumor Assays , bcl-X Protein/metabolism
3.
J Steroid Biochem Mol Biol ; 117(1-3): 23-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19563893

ABSTRACT

Superoxide dismutase (SOD) occurs in two intracellular forms in mammals, copper-zinc SOD (CuZnSOD), found in the cytoplasm, mitochondria and nucleus, and manganese superoxide dismutase (MnSOD), in mitochondria. Changes in MnSOD expression (as compared to normal cells) have been reported in several forms of cancer, and these changes have been associated with regulation of cell proliferation, cell death, and metastasis. We have found that progestins stimulate MnSOD in T47D human breast cancer cells in a time and physiological concentration-dependent manner, exhibiting specificity for progestins and inhibition by the antiprogestin RU486. Progestin stimulation occurs at the level of mRNA, protein, and enzyme activity. Cycloheximide inhibits stimulation at the mRNA level, suggesting that progestin induction of MnSOD mRNA depends on synthesis of protein. Experiments with the MEK inhibitor UO126 suggest involvement of the MAP kinase signal transduction pathway. Finally, MnSOD-directed siRNA lowers progestin-stimulated MnSOD and inhibits progestin stimulation of migration and invasion, suggesting that up-regulation of MnSOD may be involved in the mechanism of progestin stimulation of invasive properties. To our knowledge, this is the first characterization of progestin stimulation of MnSOD in human breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Progesterone Congeners/pharmacology , Superoxide Dismutase/metabolism , Base Sequence , Breast Neoplasms/etiology , Butadienes/pharmacology , Cell Line, Tumor , DNA Primers/genetics , Female , Hormone Antagonists/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Mifepristone/pharmacology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , Nitriles/pharmacology , Promegestone/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Receptors, Progesterone/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics
4.
Am J Pathol ; 168(5): 1608-18, 2006 May.
Article in English | MEDLINE | ID: mdl-16651627

ABSTRACT

Alzheimer's disease is a multifactorial, progressive, age-related neurodegenerative disease. In familial Alzheimer's disease, Abeta is excessively produced and deposited because of mutations in the amyloid precursor protein, presenilin-1, and presenilin-2 genes. Here, we generated a double homozygous knock-in mouse model that incorporates the Swedish familial Alzheimer's disease mutations and converts mouse Abeta to the human sequence in amyloid precursor protein and had the P264L familial Alzheimer's disease mutation in presenilin-1. We observed Abeta deposition in double knock-in mice beginning at 6 months as well as an increase in the levels of insoluble Abeta1-40/1-42. Brain homogenates from 3-, 6-, 9-, 12-, and 14-month-old mice showed that protein levels of manganese superoxide dismutase (MnSOD) were unchanged in the double knock-in mice compared to controls. Genotype-associated increases in nitrotyrosine levels were observed. Protein immunoprecipitation revealed MnSOD as a target of this nitration. Although the levels of MnSOD protein did not change, MnSOD activity and mitochondrial respiration decreased in knock-in mice, suggesting compromised mitochondrial function. The compromised activity of MnSOD, a primary antioxidant enzyme protecting mitochondria, may explain mitochondrial dysfunction and provide the missing link between Abeta-induced oxidative stress and Alzheimer's disease.


Subject(s)
Aging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Oxidative Stress/physiology , Superoxide Dismutase/metabolism , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Brain/metabolism , Cerebral Cortex/metabolism , Homozygote , Mice , Mice, Mutant Strains , Mice, Transgenic , Mitochondria/physiology , Respiration , Tyrosine/analogs & derivatives , Tyrosine/metabolism
5.
J Steroid Biochem Mol Biol ; 98(4-5): 218-27, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16466914

ABSTRACT

Previously, we have shown that progestins both stimulate proliferation of the progesterone receptor (PR)-rich human breast cancer cell line T47D and protect from cell death, in charcoal-stripped serum-containing medium. To lessen the variability inherent in different preparations of serum, we decided to further characterize progestin inhibition of cell death using serum starvation to kill the cells, and find that progestins protect from serum-starvation-induced apoptosis in T47D cells. This effect exhibits specificity for progestins and is inhibited by the antiprogestin RU486. While progestin inhibits cell death in a dose-responsive manner at physiological concentrations, estradiol-17beta surprisingly does not inhibit cell death at any concentration from 0.001 nM to 1 microM. Progestin inhibition of cell death also occurs in at least two other human breast cancer cell lines, one with an intermediate level of PR, MCF-7 cells, and, surprisingly, one with no detectable level of PR, MDA-MB-231 cells. Further, we have found progestin inhibition of cell death caused by the breast cancer chemotherapeutic agents doxorubicin and 5-fluorouracil. These data are consistent with the building body of evidence that progestins are not the benign hormones for breast cancer they have been so long thought to be, but may be harmful both for undiagnosed cases and those undergoing treatment.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Progestins/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Estradiol/pharmacology , Female , Fluorouracil/pharmacology , Humans , Receptors, Progesterone/metabolism , Tumor Cells, Cultured
6.
J Mol Cell Cardiol ; 39(5): 792-803, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16140321

ABSTRACT

Tamoxifen (TAM), a synthetic nonsteroidal antiestrogen effectively and widely used for breast cancer treatment, is known to have antioxidant and cardioprotective effects, but whether the beneficial cardiovascular effect of TAM is linked to its antioxidant effect is unknown. In this study, we investigated the effect of TAM on the levels of manganese superoxide dismutase (MnSOD), a mitochondrial antioxidant enzyme, in cardiac tissues and cardiomyocytes. TAM treatment induced MnSOD expression in vitro and in vivo. Cardiomyocytes isolated from TAM-pretreated mice also had higher MnSOD levels and fewer apoptotic cells compared to cardiomyocytes from control mice after adriamycin (ADR) treatment. To further confirm the role of MnSOD in the protection against ADR in cardiomyocytes, we used cardiomyocytes isolated from MnSOD knock-out (MnSOD(+/-)), wild-type (NTg) and human MnSOD transgenic (TgH) mice. TUNEL assay indicated that the percentage of cells undergoing apoptosis after ADR treatment was significantly greater in MnSOD(+/-) than in NTg or TgH cardiomyocytes. 3-[4, 5-Dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay showed that basal level of mitochondrial function was lower in MnSOD(+/-) cardiomyocytes than in NTg or TgH, and that MnSOD(+/-) was more sensitive to ADR. ADR treatment increased caspase activity, which was significantly higher in MnSOD(+/-) than in NTg or TgH cardiomyocytes. These results suggested that TAM-induced MnSOD expression is at least, in part, contribute to the cardioprotective effects of TAM.


Subject(s)
Cardiotonic Agents/pharmacology , Superoxide Dismutase/metabolism , Tamoxifen/pharmacology , Animals , Apoptosis , Cardiotonic Agents/therapeutic use , Caspase Inhibitors , Caspases/metabolism , Doxorubicin/pharmacology , Enzyme Induction , Formazans/pharmacology , Gene Expression , Genotype , In Situ Nick-End Labeling , Male , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tamoxifen/therapeutic use , Tetrazolium Salts/pharmacology
7.
Biochem J ; 384(Pt 3): 543-9, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15330761

ABSTRACT

Cytokines, phorbol esters, radiation and chemotherapeutic drugs up-regulate the expression of MnSOD (manganese superoxide dismutase). Using the VA-13 cell line, we studied the regulation of SOD2 upon treatment with PMA. Pre-treatment with CHX (cycloheximide) followed by PMA led to significantly higher levels of MnSOD mRNA compared with those with either agent alone, suggesting de novo synthesis of an inhibitory protein. PMA treatment modulates redox-sensitive transcription factors, therefore we evaluated the effects of this combination treatment upon AP-1 (activator protein 1) and NF-kappaB (nuclear factor kappaB), two trans-acting factors suggested to play a role in SOD2 regulation. Co-administration of CHX and PMA led to a time-dependent increase in the binding activity of NF-kappaB. Therefore we evaluated IkappaBalpha (inhibitory kappaBalpha) and found that co-administration decreased its steady-state level compared with either agent alone, suggesting that enhanced NF-kappaB activation is due to inhibition of IkappaBalpha synthesis. PMA activates PKC (protein kinase C) enzymes which phosphorylate IkappaBalpha, leading to its degradation, therefore we used GF109203X to inhibit PKC activity. Stable transfection utilizing a PMA-responsive element in the human SOD2 gene, showed a concentration-dependent decrease in luciferase and NF-kappaB-binding activity with GF109203X. Western blot analysis indicated the presence of several PKC isoforms in the VA-13 cell line; however, PMA pre-treatment specifically down-regulated alpha and betaI, suggesting a role for one or more of these proteins in SOD2 induction. Taken together, these results indicate that the PKC pathway leading to SOD2 induction proceeds at least in part through NF-kappaB and that inhibition of IkappaBalpha synthesis might serve as a potential pharmacological approach to up-regulate MnSOD.


Subject(s)
Down-Regulation , Gene Expression Regulation, Enzymologic , I-kappa B Proteins/metabolism , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Cell Line , Cycloheximide/pharmacology , DNA/metabolism , Down-Regulation/drug effects , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , Indoles/pharmacology , Introns/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Maleimides/pharmacology , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Protein Binding/drug effects , Protein Biosynthesis/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Superoxide Dismutase/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transcription Factor AP-1/metabolism
8.
Antioxid Redox Signal ; 6(3): 537-48, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15130280

ABSTRACT

To study early subcellular pathologic changes of tumorigenesis in mouse skin and possible modulation by overexpression of the mitochondrial antioxidant enzyme manganese superoxide dismutase (MnSOD), skin keratinocytes from nontransgenic (Ntg) and transgenic (TgH) mice overexpressing MnSOD topically treated with one dose of 7,12-dimethylbenz(a)anthracene (DMBA) and a subsequent dose of 12-O-tetradecanoylphorbol 13-acetate (TPA) were analyzed in situ for levels of MnSOD and the oxidative damage product 4-hydroxy-2-nonenal (4HNE)-modified proteins using specific antibodies and immunogold electron microscopy. At all selected time points analyzed after TPA treatment, there was more MnSOD immunoreactive protein in mitochondria of keratinocytes of TgH mice than Ntg mice. Compared with untreated groups, there was a large increase in 4HNE-modified proteins at 6-24 h after TPA treatment, and this increase was larger in Ntg than TgH mice. Indices of mitosis and apoptosis of keratinocytes were greater in DMBA/TPA-treated TgH than Ntg mouse skin. Mitochondrial injury detected by transmission electron microscopy was delayed in keratinocytes of TgH compared with Ntg mice. The present study demonstrated that overexpression of MnSOD not only protected cells from oxidative damage, but also affected cell turnover kinetics. Thus, previously identified reduction in papilloma formation observed in TgH mice is correlated with mitochondrial events.


Subject(s)
Mitochondria/pathology , Oxygen/metabolism , Skin Neoplasms/metabolism , Superoxide Dismutase/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA, Complementary/metabolism , Female , Humans , Immunohistochemistry , Keratinocytes/metabolism , Kinetics , Mice , Mice, Transgenic , Microscopy, Electron , Mitochondria/metabolism , Signal Transduction , Skin/pathology , Skin/ultrastructure , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
9.
Oncogene ; 21(22): 3603-10, 2002 May 16.
Article in English | MEDLINE | ID: mdl-12032862

ABSTRACT

Manganese superoxide dismutase (MnSOD) has been shown to suppress the development of cancer. Tamoxifen (TAM), a nonsteroidal anti-estrogen that is widely used in chemotherapy, is known to be a modulator of antioxidant status. However, the mechanism by which TAM mediates antioxidant enzyme induction remains unclear. In this study we investigated TAM enhancement of MnSOD induction by TNF-alpha. The results show that co-treatment with TAM and TNF-alpha increases the MnSOD promoter/enhancer driven luciferase activity, MnSOD mRNA and protein levels. Interestingly, co-treatment with TAM and TNF-alpha drastically decreases the binding activity of the p50/p50 homodimer and increases that of the p50/p65 heterodimer compared to TNF-alpha alone. This change in DNA binding could not be attributed to a decrease in the level of p50, its precursor, p105, or its inhibitors. Furthermore, TAM did not enhance degradation of IkappaB-alpha. These results suggest that p50/p50 homodimer may act as an inhibitory complex of MnSOD expression. Modulation of the DNA binding activity in favor of the p50/p65 complex may enhance NF-kappaB mediated induction of MnSOD by TAM. These findings reveal a potential novel mechanism for the induction of the human MnSOD gene.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Estrogen Antagonists/pharmacology , I-kappa B Proteins , NF-kappa B/metabolism , Superoxide Dismutase/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Cell Line, Transformed , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Deoxycholic Acid/pharmacology , Dimerization , Drug Synergism , Enhancer Elements, Genetic , Humans , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit , RNA, Messenger/biosynthesis , Superoxide Dismutase/biosynthesis , Transcriptional Activation
10.
Methods Enzymol ; 349: 306-12, 2002.
Article in English | MEDLINE | ID: mdl-11912921

ABSTRACT

The human MnSOD gene has a typical housekeeping gene promoter, but is highly inducible by various physical, chemical, and biological agents. Transcription factors SP-1 and AP-2 seem to have opposite roles in the transcriptional activity of the basal promoter. Whereas SP-1 plays a positive role, which is absolutely essential for transcription from the human MnSOD promoter, AP-2 appears to play a negative role in this process. An enhancer element is found in the promoter region of the human MnSOD gene. Several important enhancer elements are located in the second intron. The NF-kappa B site in the second intron is essential but not sufficient for high-level induction of MnSOD by cytokines. Although mutations in the regulatory elements may be partially responsible for the lack of induction of MnSOD in some cell types, differences in the degree of induction exist that cannot be accounted for by the defect in the DNA sequence. It is highly likely that this difference is due to the presence or absence of coactivator or suppressor proteins in the cells and may have a physiological role in the defense against oxidative stress.


Subject(s)
Superoxide Dismutase/genetics , Base Sequence , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Humans , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sp1 Transcription Factor/metabolism , Superoxide Dismutase/biosynthesis , Transcription Factor AP-2 , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...