Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Telemed Telecare ; 26(4): 189-199, 2020 May.
Article in English | MEDLINE | ID: mdl-30541375

ABSTRACT

AIMS: The purpose of this research was to investigate the effectiveness of telemonitoring for chronic obstructive pulmonary disease. METHODS: We searched MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials and CINAHL up to September 2018. We selected randomised controlled trials comparing telemonitoring and control groups for chronic obstructive pulmonary disease management. Two reviewers independently examined articles based on eligibility, extracted data and evaluated the risk of bias. The Cochrane tool was applied for assessing the risk of bias. The 95% confidence interval was calculated. RESULTS: A total of 28 randomised controlled trials were included. Meta-analysis revealed that there were no variables showing a statistically significant difference between telemonitoring and control groups. Chronic obstructive pulmonary disease exacerbation rate (six studies) was not different between two groups (risk ratio 0.67, 95% confidence interval 0.31-1.42). Subgroup analysis showed that telemonitoring reduced exacerbation rates when the intervention continued for longer than six months or pulmonary function was monitored. No differences between groups were noticed for mortality (seven studies, risk ratio 0.89, 95% confidence interval 0.60-1.34). Similarly, no differences between groups were observed in the patient-reported outcomes (St George's Respiratory Questionnaire, Chronic Respiratory Disease Questionnaire-Dyspnea score) and for health service utilization (length of hospital stay, number of hospital admissions, number of emergency room visits). CONCLUSIONS: Telemonitoring for chronic obstructive pulmonary disease was unlikely to result in statistically significant improvements in health outcomes. However, our novel finding was that at least six months of intervention duration and monitoring of pulmonary function play roles in activating the effects of telemonitoring.


Subject(s)
Decision Support Systems, Clinical , Patient Discharge/statistics & numerical data , Pulmonary Disease, Chronic Obstructive/therapy , Telemedicine/methods , Telemetry/methods , Hospitalization/statistics & numerical data , Humans , Length of Stay/statistics & numerical data , Quality of Life , Quality-Adjusted Life Years , Randomized Controlled Trials as Topic
2.
PLoS One ; 9(6): e99047, 2014.
Article in English | MEDLINE | ID: mdl-24964199

ABSTRACT

Swedish double mutation (KM670/671NL) of amyloid precursor protein (APP) is reported to increase toxic amyloid ß (Aß) production via aberrant cleavage at the ß-secretase site and thereby cause early-onset Alzheimer's disease (AD). However, the underlying molecular mechanisms leading to AD pathogenesis remains largely unknown. Previously, our transcriptome sequence analyses revealed global expressional modifications of over 600 genes in APP-Swedish mutant-expressing H4 (H4-sw) cells compared to wild type H4 cells. Insulin-like growth factor binding protein 3 (IGFBP3) is one gene that showed significantly decreased mRNA expression in H4-sw cells. In this study, we investigated the functional role of IGFBP3 in AD pathogenesis and elucidated the mechanisms regulating its expression. We observed decreased IGFBP3 expression in the H4-sw cell line as well as the hippocampus of AD model transgenic mice. Treatment with exogenous IGFBP3 protein inhibited Aß1-42- induced cell death and caspase-3 activity, whereas siRNA-mediated suppression of IGFBP3 expression induced cell death and caspase-3 cleavage. In primary hippocampal neurons, administration of IGFBP3 protein blocked apoptotic cell death due to Aß1-42 toxicity. These data implicate a protective role for IGFBP3 against Aß1-42-mediated apoptosis. Next, we investigated the regulatory mechanisms of IGFBP3 expression in AD pathogenesis. We observed abnormal IGFBP3 hypermethylation within the promoter CpG island in H4-sw cells. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine restored IGFBP3 expression at both the mRNA and protein levels. Chronic exposure to Aß1-42 induced IGFBP3 hypermethylation at CpGs, particularly at loci -164 and -173, and subsequently suppressed IGFBP3 expression. Therefore, we demonstrate that expression of anti-apoptotic IGFBP3 is regulated by epigenetic DNA methylation, suggesting a mechanism that contributes to AD pathogenesis.


Subject(s)
Amyloid beta-Peptides/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Caspase 3/metabolism , Cell Survival/genetics , CpG Islands , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation , Humans , Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 3/genetics , Mice , Mice, Transgenic , Rats
3.
Oncol Rep ; 32(2): 815-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920423

ABSTRACT

The metastatic properties of cancer cells result from genetic and epigenetic alterations that lead to the abnormal expression of key genes regulating tumor phenotypes. Recent discoveries suggest that aberrant DNA methylation provides cancer cells with advanced metastatic properties; however, the precise regulatory mechanisms controlling metastasis-associated genes and their roles in metastatic transformation are largely unknown. We injected SK-OV-3 human ovarian cancer cells into the perineum of nude mice to generate a mouse model that mimics human ovarian cancer metastasis. We analyzed the mRNA expression and DNA methylation profiles in metastasized tumor tissues in the mice. The pro-oncogenic anterior gradient 2 (AGR2) gene showed increased mRNA expression and hypomethylation at CpG sites in its promoter region in the metastatic tumor tissues compared with the cultured SK-OV-3 cells. We identified crucial cytosine residues at CpG sites in the AGR2 promoter region. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine reduced the level of CpG methylation in the AGR2 promoter and increased the level of AGR2 expression. Next, we explored the functional role of AGR2 in the metastatic transformation of SK-OV-3 cells. SK-OV-3 cells overexpressing AGR2 showed increased migratory and invasive activity. Our results indicate that DNA methylation within the AGR2 promoter modulates more aggressive cancer cell phenotypes.


Subject(s)
DNA Methylation , Neoplasm Metastasis/pathology , Ovarian Neoplasms/pathology , Proteins/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Decitabine , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred BALB C , Mucoproteins , Neoplasm Metastasis/genetics , Neoplasms, Experimental , Oligonucleotide Array Sequence Analysis , Oncogene Proteins , Ovarian Neoplasms/genetics , Promoter Regions, Genetic
4.
Oncol Rep ; 31(5): 2139-46, 2014 May.
Article in English | MEDLINE | ID: mdl-24676393

ABSTRACT

A lack of reliable biomarkers for the early detection and risk of metastatic recurrences makes ovarian cancer the most lethal gynecological cancer. To understand the molecular mechanisms involved in ovarian cancer metastasis in vivo, we analyzed the transcriptional expression pattern in metastatic implants of human ovarian carcinoma xenografts in mice. The expression of 937 genes was significantly different, by at least 2-fold, in the xenografts compared with that in SK-OV-3 cells. We investigated the mechanisms that regulate the expression of one of the profoundly upregulated genes, interferon-induced transmembrane protein 1 (IFITM1), in the metastatic implants. Specific CpG sites within the IFITM1 promoter were hypomethylated in the metastatic implants relative to those in the wild-type SK-OV-3 cells. Treating wild-type SK-OV-3 cells with the demethylating agent 5-aza-2'-deoxycytidine enhanced IFITM1 expression in a dose-dependent manner, implying transcriptional regulation by promoter methylation. We also found that IFITM1 overexpression caused increased migration and invasiveness in SK-OV-3 cells. Our results demonstrate that IFITM1 could be a novel metastasis-promoting gene that enhances the metastatic phenotype in ovarian cancer via epigenetic transcriptional regulation. Our findings also suggest that the status of DNA methylation within the IFITM1 promoter region could be a biomarker indicating metastatic progression in ovarian cancer.


Subject(s)
Antigens, Differentiation/genetics , DNA Methylation/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Biomarkers, Tumor , Cell Line, Tumor , Decitabine , Female , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/genetics , Ovary/pathology , Promoter Regions, Genetic , Xenograft Model Antitumor Assays
5.
Neurochem Res ; 36(11): 1991-2001, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21688047

ABSTRACT

Dopamine (DA), as a neurotoxin, can elicit severe Parkinson's disease-like syndrome by elevating intracellular reactive oxygen species (ROS) levels and apoptotic activity. We examined the inhibitory effects of 3α-acetoxyeudesma-1,4(15),11(13)-trien-12,6α-olide (AETO), purified from the leaves of Laurus nobilis L., on DA-induced apoptosis and α-synuclein (α-syn) formation in dopaminergic SH-SY5Y cells. AETO decreased the active form of caspase-3 and the levels of p53, which were accompanied by increased levels of Bcl-2 in a dose-dependent manner. Flow cytometric and Western blot analysis showed that AETO significantly inhibited DA-induced apoptosis along with suppression of intracellular tyrosinase activity, ROS generation, quinoprotein, and α-syn formation (P < 0.01). These results indicate that AETO inhibited DA-induced apoptosis, which is closely related to the suppression of intracellular tyrosinase activity and the formation of α-syn, ROS, and quinoprotein in SH-SY5Y cells.


Subject(s)
Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Sesquiterpenes, Eudesmane/therapeutic use , Antioxidants/therapeutic use , Caspase 3/metabolism , Cell Line, Tumor , Dopamine/pharmacology , Humans , Laurus/chemistry , Monophenol Monooxygenase/antagonists & inhibitors , Neuroblastoma , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , alpha-Synuclein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...