Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Public Health (Oxf) ; 43(4): e632-e636, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34918088

ABSTRACT

BACKGROUND: The purpose of this study is to investigate the drinking behavior of Korean pregnant women in 2017 and to compare the changes of drinking status with the results of the research conducted in 1997 and 2008. METHODS: Pregnant women at one obstetrics and gynecology hospital and one university hospital were the subjects of the study. They were filled out questionnaire. RESULTS: The rate of positive responses to CAGE increased 16.0% in 2008 and 16.3% in 2017 compared to 11.8% in 1997 (P = 0.046). Blackout history rate was keep increasing from 1997 to 2017 (8.8% versus 27.7% versus 36.3%, P < 0.001). The rate of family history of alcohol was nearly doubled in 2017 (30.3%) compared to 1997 (17.6%) and 2008 (16.3) (P < 0.001). The rate of pregnant women who drink alcohol during pregnancy decreased from 57.5% in 1997 to 39.5% in 2008 and decreased to 25.6% in 2017 (P < 0.001). The rate of pregnant women who drink alcohol after knowing the pregnancy was decreased in 2017 (6.9%) compared to 2008 (23.5%) (P < 0.001). CONCLUSION: According to the results of the study in 2017, the rate of pregnant women who drink alcohol after pregnancy was decreased compared to 1997 and 2008. However drinking behavior severity has increased in 2017.


Subject(s)
Alcohol Drinking , Pregnant Women , Alcohol Drinking/epidemiology , Female , Health Behavior , Humans , Pregnancy , Republic of Korea/epidemiology , Surveys and Questionnaires
2.
Int J Cancer ; 143(8): 1978-1993, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29744876

ABSTRACT

Tumor metastasis remains the cause of 90% of cancer-related deaths. Cancer stem cells (CSC) are thought to be responsible for the aggressive and metastatic nature of triple-negative breast cancers (TNBC), and new therapeutic strategies are being devised to target them. Flubendazole (FLU) is a widely used anthelmintic agent that also exhibits anticancer activity in several cancer types. The aim of this study was to characterize the mechanism of action of FLU on breast cancer stem cell (BCSC)-like properties and metastasis in TNBC. FLU treatment caused a significant induction of apoptosis, accompanied by G2/M phase accumulation, caspase-3/-7 activation and the dysregulation of STAT3 activation in TNBC cells. The latter phenomenon was associated with impairment of cancer stem-like traits, concomitant with a reduction in the CD24low /CD44high , CD24high /CD49fhigh subpopulation, ALDH1 activity and mammosphere formation. The BCSC-enriched populations exhibited enhanced metastasis with higher STAT3 activation, while FLU administration inhibited tumor growth, angiogenesis and lung and liver metastasis, coinciding with decreased MMP-2 and MMP-9 levels in circulating blood. FLU kills not only rapid proliferating tumor cells but also effectively eradicates BCSC-like cells in vitro and in vivo. Our findings warrant further investigation of FLU as a treatment for metastatic TNBC.


Subject(s)
Mebendazole/analogs & derivatives , Neoplasm Metastasis/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Antigens, CD/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Mebendazole/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism
3.
Cell Signal ; 36: 230-239, 2017 08.
Article in English | MEDLINE | ID: mdl-28499884

ABSTRACT

Ubiquitin-specific protease 34 (USP34) is a deubiquitinating enzyme that regulates Axin stability and plays a critical role in Wnt/ß-catenin signaling. We sought to investigate the role of USP34 on epithelial-mesenchymal (EMT) induction and its effects on mammary epithelial stem cells. USP34 expression levels were relatively lower in MDA-MB-231 and 4T1 mesenchymal-like cells when compared to epithelial-like cells. Inhibition of USP34 in NMuMG cells induced EMT, as evidenced by the upregulation of EMT markers including N-cadherin, phospho-Smad3, Snail and active-ß-catenin, as well as the downregulation of Axin 1 and E-cadherin. USP34 knockdown (KD) in these cells also resulted in the acquisition of invasive behavior, and promoted stemness as indicated by enhanced mammosphere-forming ability, concomitant with the upregulation of Nanog, Oct4 and Sox2 mRNA expression. Endogenous USP34 expression was observed to be at low levels in virgin mouse mammary glands in vivo. When USP34-KD cells were transplanted into the cleared mammary fat pads (CFP) of mice, these cells reconstituted the mammary gland with ductal tree development within 3months. Our findings suggest a previously unknown role for USP34 in mammary gland development.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Stem Cells/metabolism , Ubiquitin-Specific Proteases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Self Renewal/drug effects , Down-Regulation/drug effects , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Humans , Mice , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Stem Cells/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Ubiquitin-Specific Proteases/metabolism
4.
Cancer Lett ; 386: 151-160, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27894956

ABSTRACT

Triple-negative breast cancers (TNBC) often exhibit an aggressive phenotype. Disulfiram (DSF) is an approved drug for the treatment of alcohol dependence, but has also been shown to kill TNBC cells in a copper (Cu)-dependent manner. Exactly how this occurs has not been clearly elucidated. We sought to investigate the mechanisms responsible for DSF/Cu-dependent induction of apoptosis and suppression of lung colonization by TNBC cells. DSF/Cu induced anoikis and significantly suppressed cell migration and invasion with negative effects on focal adhesions, coinciding with vimentin breakdown and calpain activation in TNBC cells. In a xenograft tumor model, DSF suppressed tumor growth and lung nodule growth, which was also associated with calpain activation. These findings warrant further investigation of disulfiram as a potential treatment for metastatic TNBC.


Subject(s)
Anoikis/drug effects , Antineoplastic Agents/pharmacology , Calpain/metabolism , Cell Movement/drug effects , Disulfiram/pharmacology , Lung Neoplasms/prevention & control , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Copper/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/enzymology , Cytoskeleton/pathology , Dose-Response Relationship, Drug , Enzyme Activation , Female , Focal Adhesions/drug effects , Focal Adhesions/enzymology , Focal Adhesions/pathology , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Proteolysis , Signal Transduction/drug effects , Time Factors , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Vimentin/metabolism , Xenograft Model Antitumor Assays
5.
Cancer Lett ; 379(1): 39-48, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27238567

ABSTRACT

HER2-positive breast tumors are known to harbor cancer stem-like cell populations and are associated with an aggressive tumor phenotype and poor clinical outcomes. Disulfiram (DSF), an anti-alcoholism drug, is known to elicit cytotoxicity in many cancer cell types in the presence of copper (Cu). The objective of the present study was to investigate the mechanism of action responsible for the induction of apoptosis by DSF/Cu and its effect on cancer stem cell properties in HER2-positive breast cancers in vitro and in vivo. DSF/Cu treatment induced apoptosis, associated with a marked decrease in HER2, truncated p95HER2, phospho-HER2, HER3, phospho-HER3 and phospho-Akt levels, and p27 nuclear accumulation. This was accompanied by the eradication of cancer stem-like populations, concomitant with the suppression of aldehyde dehydrogenase 1 (ALDH1) activity and mammosphere formation. DSF administration resulted in a significant reduction in tumor growth and an enhancement of apoptosis, as well as HER2 intracellular domain (ICD) and ALDH1A1 downregulation. Our results demonstrate that DSF/Cu induces apoptosis and eliminates cancer stem-like cells via the suppression of HER2/Akt signaling, suggesting that DSF may be potentially effective for the treatment of HER2-positive cancers.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Copper/pharmacology , Disulfiram/pharmacology , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Phenotype , Phosphorylation , Retinal Dehydrogenase , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Biochim Biophys Acta ; 1863(6 Pt A): 1071-81, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26975580

ABSTRACT

The angiotensin II type I receptor (AGTR1) has been implicated in diverse aspects of human disease, from the regulation of blood pressure and cardiovascular homeostasis to cancer progression. We sought to investigate the role of AGTR1 in cell proliferation, epithelial-mesenchymal transition (EMT), migration, invasion, angiogenesis and tumor growth in the breast cancer cell line MCF7. Stable overexpression of AGTR1 was associated with accelerated cell proliferation, concomitant with increased expression of survival factors including poly(ADP-ribose) polymerase (PARP) and X-linked inhibitor of apoptosis (XIAP), as well as extracellular signal-regulated kinase (ERK) activation. AGTR1-overexpressing MCF7 cells were more aggressive than their parent line, with significantly increased activity in migration and invasion assays. These observations were associated with changes in EMT markers, including reduced E-cadherin expression and increased p-Smad3, Smad4 and Snail levels. Treatment with the AGTR1 antagonist losartan attenuated these effects. AGTR1 overexpression also accelerated tumor growth and increased Ki-67 expression in a xenograft model. This was associated with increased tumor angiogenesis, as evidenced by a significant increase in microvessels in the intratumoral and peritumoral areas, and enhanced tumor invasion, with the latter response associated with increased EMT marker expression and matrix metallopeptidase 9 (MMP-9) upregulation. In vivo administration of losartan significantly reduced both tumor growth and angiogenesis. Our findings suggest that AGTR1 plays a significant role in tumor aggressiveness, and its inhibition may have therapeutic implications.


Subject(s)
Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Neovascularization, Pathologic/genetics , Receptor, Angiotensin, Type 1/genetics , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blotting, Western , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Losartan/pharmacology , MCF-7 Cells , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Receptor, Angiotensin, Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transplantation, Heterologous , Tumor Burden/drug effects , Tumor Burden/genetics
7.
PLoS One ; 10(11): e0141919, 2015.
Article in English | MEDLINE | ID: mdl-26528725

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive tumor subtype with an enriched CD44+/CD24- stem-like population. Salinomycin is an antibiotic that has been shown to target cancer stem cells (CSC); however, the mechanisms of action involved have not been well characterized. The objective of the present study was to investigate the effect of salinomycin on cell death, migration, and invasion, as well as CSC-like properties in MDA-MB-231 breast cancer cells. Salinomycin significantly induced anoikis-sensitivity, accompanied by caspase-3 and caspase-8 activation and PARP cleavage, during anchorage-independent growth. Salinomycin treatment also caused a marked suppression of cell migration and invasion with concomitant downregulation of MMP-9 and MMP-2 mRNA levels. Notably, salinomycin inhibited the formation of mammospheres and effectively reduced the CD44+/CD24- stem-like population during anchorage-independent growth. These observations were associated with the inhibition of STAT3 phosphorylation (Tyr705). Furthermore, interleukin-6 (IL-6)-induced STAT3 activation was strongly suppressed by salinomycin challenge. These findings support the notion that salinomycin may be potentially efficacious for targeting breast cancer stem-like cells through the inhibition of STAT3 activation.


Subject(s)
Breast Neoplasms/metabolism , Hyaluronan Receptors , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Pyrans/pharmacology , STAT3 Transcription Factor/metabolism , Sialic Acid Binding Ig-like Lectin 2 , Anoikis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , STAT3 Transcription Factor/genetics
8.
Biochem Biophys Res Commun ; 466(4): 696-703, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26407842

ABSTRACT

Cancer stem cells (CSCs) play important roles in the formation, growth and recurrence of tumors, particularly following therapeutic intervention. Salinomycin has received recent attention for its ability to target breast cancer stem cells (BCSCs), but the mechanisms of action involved are not fully understood. In the present study, we sought to investigate the mechanisms responsible for salinomycin's selective targeting of BCSCs and its anti-tumor activity. Salinomycin suppressed cell viability, concomitant with the downregulation of cyclin D1 and increased p27(kip1) nuclear accumulation. Mammosphere formation assays revealed that salinomycin suppresses self-renewal of ALDH1-positive BCSCs and downregulates the transcription factors Nanog, Oct4 and Sox2. TUNEL analysis of MDA-MB-231-derived xenografts revealed that salinomycin administration elicited a significant reduction in tumor growth with a marked downregulation of ALDH1 and CD44 levels, but seemingly without the induction of apoptosis. Our findings shed further light on the mechanisms responsible for salinomycin's effects on BCSCs.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Pyrans/pharmacology , Aldehyde Dehydrogenase 1 Family , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation/drug effects , Female , Humans , Hyaluronan Receptors/metabolism , Isoenzymes/antagonists & inhibitors , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Retinal Dehydrogenase/antagonists & inhibitors , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL