Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
BMC Med Educ ; 24(1): 101, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38291422

ABSTRACT

BACKGROUND: The aging population has caused assistive technology (AT) to receive attention. Thus, ensuring accurate user comprehension of AT has become increasingly crucial, and more specialized education for students in relevant fields is necessary. The goal of this study was to explore the learning outcomes in the context of AT for older adults and individuals with disabilities through the use of VR experiential learning. METHODS: A parallel-group design was used. Sixty third-year university students studying gerontology and long-term-care-related subjects in Taiwan were enrolled, with the experimental (VR) and control (two-dimensional [2D] video) groups each comprising 30 participants. Both groups received the same 15-minute lecture. Subsequently, the experimental group received experiential learning through a VR intervention, whereas the control group watched a 2D video to learn. The students' knowledge of AT was assessed using a pretest and posttest. Additionally, their skills in evaluation of residential environments were assessed using the Residential Environment Assessment (REA) Form for Older Adults. All data analyses were performed with SPSS version 22. RESULTS: In the posttest conducted after the intervention, the experimental group exhibited a significant 20.67 point improvement (p < 0.05), whereas the control group only exhibited improvement of 3.67 points (p = 0.317). Furthermore, the experimental group demonstrated a significantly higher score (+ 2.17 points) on the REA Form for Older Adults than did the control group (p < 0.05). CONCLUSION: VR experiential learning can significantly improve undergraduate students' knowledge and evaluation skills in relation to AT for older adults and individuals with disabilities.


Subject(s)
Disabled Persons , Self-Help Devices , Virtual Reality , Humans , Aged , Problem-Based Learning , Students
2.
Clin Pharmacol Ther ; 114(4): 914-921, 2023 10.
Article in English | MEDLINE | ID: mdl-37463101

ABSTRACT

Taiwan's National Health Insurance (NHI) program forced discontinuation of biologic use in Crohn's disease (CD) after a limited treatment duration, regardless of disease activity. This study investigated the retreatment rate and suboptimal outcomes (i.e., CD-related surgeries, hospitalizations, emergency room visits, and oral steroid flare-ups) after forced discontinuation. This retrospective cohort study was conducted using data from the NHI Database. Patients who received ≥40 weeks of biologic treatment followed by a forced discontinuation were included. The time of biologic retreatment and the cumulative incidence of suboptimal outcomes after the forced discontinuation as well as related risk factors were analyzed. Included were 215 patients (68% male). At the beginning of biologic therapy, the mean age (±SD) was 35.7 (±13.5) years, and the disease duration was 4.46 (±3.52) years. The median (interquartile range) biologic treatment duration was 57.86 (50.3-83.3) weeks. Within the first year after forced discontinuation, 67% of patients (n = 144) were retreated with a second course of biologics, and 53% of patients (n = 114) experienced at least one suboptimal outcome. The independent risk factors associated with the occurrence of suboptimal outcomes were CD-related emergency room visits and hospitalizations during biologic therapy (hazard ratio: 2.49; 95% confidence interval: 1.59-3.89). More than two-thirds of patients with CD required biological retreatment within 1 year after a forced discontinuation. The substantial proportion of patients with poor disease outcomes highlights the need to continue the biologic.


Subject(s)
Biological Products , Crohn Disease , Humans , Male , Young Adult , Adult , Middle Aged , Female , Crohn Disease/diagnosis , Crohn Disease/drug therapy , Retrospective Studies , Biological Factors , Retreatment , Biological Products/adverse effects
3.
Inflamm Bowel Dis ; 29(11): 1730-1740, 2023 11 02.
Article in English | MEDLINE | ID: mdl-36626567

ABSTRACT

BACKGROUND: This nationwide prospective registry study investigated the real-world effectiveness, safety, and persistence of vedolizumab (VDZ) in inflammatory bowel disease (IBD) patients in Taiwan. Disease relapse rates after VDZ discontinuation due to reimbursement restriction were assessed. METHODS: Data were collected prospectively (January 2018 to May 2020) from the Taiwan Society of IBD registry. RESULTS: Overall, 274 patients (147 ulcerative colitis [UC] patients, 127 Crohn's disease [CD] patients) were included. Among them, 70.7% with UC and 50.4% with CD were biologic-naïve. At 1 year, 76.0%, 58.0%, 35.0%, and 62.2% of UC patients and 57.1%, 71.4%, 33.3%, and 30.0% of CD patients achieved clinical response, clinical remission, steroid-free remission, and mucosal healing, respectively. All patients underwent hepatitis B and tuberculosis screening before initiating biologics, and prophylaxis was recommended when necessary. One hepatitis B carrier, without antiviral prophylaxis due to economic barriers, had hepatitis B reactivation during steroid tapering and increasing azathioprine dosage, which was controlled with an antiviral agent. No tuberculosis reactivation was noted. At 12 months, non-reimbursement-related treatment persistence rates were 94.0% and 82.5% in UC and CD patients, respectively. Moreover, 75.3% of IBD patients discontinued VDZ due to mandatory drug holiday. Relapse rates after VDZ discontinuation at 6 and 12 months were 36.7% and 64.3% in CD patients and 42.9% and 52.4% in UC patients, respectively. CONCLUSIONS: The findings demonstrated VDZ effectiveness in IBD patients in Taiwan, with high treatment persistence rates and favorable safety profiles. A substantial IBD relapse rate was observed in patients who had mandatory drug holiday.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Hepatitis B , Inflammatory Bowel Diseases , Humans , Taiwan , Remission Induction , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Recurrence , Treatment Outcome , Retrospective Studies
4.
Life Sci ; 309: 121023, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36202175

ABSTRACT

AIMS: Glioblastoma multiforme (GBM) is the most aggressive and mortal primary glioma in adults. Temozolomide (TMZ) is a first-line clinical chemotherapeutic drug. However, TMZ resistance causes treatment failure in patients. Thus, exploring effective adjuvant drugs for GBM is crucial. Piperlongumine (PL), a bioactive alkaloid isolated from long pepper, possesses promising anticancer abilities. However, PL-mediated cytotoxic mechanisms in GBM are still unclear. We attempted to identify PL-regulated networks in suppressing GBM malignancy. MAIN METHODS AND KEY FINDINGS: PL treatment significantly induced more apoptotic death in several GBM cell lines than in normal astrocytes. Decreased cell invasion, colony generation, and sphere formation, and enhanced TMZ cytotoxicity were found in PL-treated cells. Through RNA sequencing, PL-mediated transcriptomic profiles were established. By intersecting PL-downregulated genes, higher expressing genes in The Cancer Genome Atlas (TCGA) tumor tissues, and risk genes in three different GBM databases, tripartite motif-containing 14 (TRIM14) was selected. Higher TRIM14 expression was correlated with poor patient survival, and it existed in tumor samples, in mesenchymal type of GBM patients, and in GBM cells. PL significantly reduced TRIM14 expression through activating the p38/MAPK pathway. Overexpression or knockdown of TRIM14 influenced cell growth, PL-inhibited cell viability, invasion, colony generation, and sphere formation. Finally, using a gene set enrichment analysis, genes positively correlated with TRIM14 levels were enriched in epithelial-to-mesenchymal transition signaling. TRIM14 overexpression attenuated PL-regulated mesenchymal transition signaling. SIGNIFICANCE: PL inhibited TRIM14 signaling through activating the p38/MAPK pathway to inhibit GBM malignancy. Our findings may provide better insights and directions for future GBM therapies.


Subject(s)
Brain Neoplasms , Dioxolanes , Glioblastoma , Humans , Temozolomide/pharmacology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Cell Line, Tumor , Dioxolanes/pharmacology , Drug Resistance, Neoplasm/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Antineoplastic Agents, Alkylating/pharmacology , Gene Expression Regulation, Neoplastic , Tripartite Motif Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
5.
Cancer Control ; 29: 10732748221124865, 2022.
Article in English | MEDLINE | ID: mdl-36134681

ABSTRACT

INTRODUCTION: While Hodgkin lymphoma (HL) is mostly curable, outcomes for advanced-stage HL remain unsatisfactory. The International Prognostic Score and its modifications were developed to predict HL prognosis; however, more straightforward prognostic factors are needed. This study aimed to identify simpler prognostic factors for advanced-stage newly diagnosed HL (NDHL). METHODS: This retrospective study used the Taiwan National Health Insurance Research Database and the Taiwan Cancer Registry. Patients with advanced-stage NDHL receiving ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) or ABVD-like regimens between 2009 and 2016 were enrolled. Cox proportional hazards models were used to identify prognostic factors for the time to next treatment (TTNT) and overall survival (OS). We used the time-dependent area under the receiver operating characteristic curve (AUROC) to evaluate model performance. RESULTS: The study included 459 patients with advanced-stage NDHL. A bimodal age distribution (peaks 20-44 and >65 years) was observed. Over a median follow-up of 4.7 years, the complete remission and OS rates were 52% and 76%, respectively. Age ≥60 years (adjusted hazard ratio [aHR]: 1.73, 95% confidence interval [CI]: 1.23-2.43), extranodal involvement (1.40, 1.05-1.87), B symptoms (1.53, 1.13-2.06), and Charlson Comorbidity Index (CCI) ≥1 (1.49, 1.08-2.06) were significantly associated with a shorter TTNT. The time-dependent AUROC was .65. With a time-dependent AUROC of .81, age ≥60 years (4.55, 2.90-7.15) and CCI ≥1 (1.86, 1.18-2.91) were risk factors for worse OS. CONCLUSION: Older age and more comorbidities were risk factors for an inferior OS in advanced-stage NDHL, while older age, extranodal involvement, B-symptoms, and higher CCI were significantly associated with disease relapse.


Subject(s)
Hodgkin Disease , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bleomycin/therapeutic use , Dacarbazine/adverse effects , Doxorubicin/therapeutic use , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Humans , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Treatment Outcome , Vinblastine/therapeutic use
6.
Cell Mol Neurobiol ; 42(3): 791-806, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33025417

ABSTRACT

The insulin-like growth factor (IGF)-1 and transforming growth factor (TGF)-ß signal pathways are both recognized as important in regulating cancer prognosis, such as the epithelial-to-mesenchymal transition (EMT) and cell invasion. However, cross-talk between these two signal pathways in glioblastoma multiforme (GBM) is still unclear. In the present study, by analyzing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GSE) 4412, GBM patients with higher IGF-1 levels exhibited poorer survival. Genes positively correlated with IGF-1 were enriched in EMT and TGF-ß signal pathways. IGF-1 treatment enhanced mesenchymal marker expressions and GBM cell invasion. A significant positive correlation was observed for IGF-1 with TGF-ß1 (TGFB1) or TGF-ß receptor 2 (TGFBR2), both of which participate in TGF-ß signaling and are risk genes in the GBM process. IGF-1 stimulation promoted both TGFB1 and TGFBR2 expressions. LY2157299, a TGF-ß signaling inhibitor, attenuated IGF-1-enhanced GBM cell invasion and mesenchymal transition. By analyzing IGF-1-regulated microRNA (miR) profiles, miR-4286 was found to be significantly downregulated in IGF-1-treated cells and could be targeted to both TGFB1 and TGFBR2. Overexpression of miR-4286 significantly attenuated expressions of the IGF-1-mediated mesenchymal markers, TGFB1 and TGFBR2. Using kinase inhibitors, only U0126 treatment showed an inhibitory effect on IGF-1-reduced miR-4286 and IGF-1-induced TGFB1/TGFBR2 expressions, suggesting that MEK/ERK signaling is involved in the IGF-1/miR-4286/TGF-ß signaling axis. Finally, our results suggested that miR-4286 might act as a tumor suppressive microRNA in inhibiting IGF-1-enhanced GBM cell invasion. In conclusion, IGF-1 is connected to TGF-ß signaling in regulating the mesenchymal transition and cell invasion of GBM through inhibition of miR-4286. Our findings provide new directions and mechanisms for exploring GBM progression.


Subject(s)
Glioblastoma , MicroRNAs , Signal Transduction , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Glioblastoma/pathology , Humans , Insulin-Like Growth Factor I/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Transforming Growth Factor beta/metabolism
7.
BMC Med ; 19(1): 59, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33627136

ABSTRACT

BACKGROUND: Long noncoding (lnc)RNAs and glycolysis are both recognized as key regulators of cancers. Some lncRNAs are also reportedly involved in regulating glycolysis metabolism. However, glycolysis-associated lncRNA signatures and their clinical relevance in cancers remain unclear. We investigated the roles of glycolysis-associated lncRNAs in cancers. METHODS: Glycolysis scores and glycolysis-associated lncRNA signatures were established using a single-sample gene set enrichment analysis (GSEA) of The Cancer Genome Atlas pan-cancer data. Consensus clustering assays and genomic classifiers were used to stratify patient subtypes and for validation. Fisher's exact test was performed to investigate genomic mutations and molecular subtypes. A differentially expressed gene analysis, with GSEA, transcription factor (TF) activity scoring, cellular distributions, and immune cell infiltration, was conducted to explore the functions of glycolysis-associated lncRNAs. RESULTS: Glycolysis-associated lncRNA signatures across 33 cancer types were generated and used to stratify patients into distinct clusters. Patients in cluster 3 had high glycolysis scores and poor survival, especially in bladder carcinoma, low-grade gliomas, mesotheliomas, pancreatic adenocarcinomas, and uveal melanomas. The clinical significance of lncRNA-defined groups was validated using external datasets and genomic classifiers. Gene mutations, molecular subtypes associated with poor prognoses, TFs, oncogenic signaling such as the epithelial-to-mesenchymal transition (EMT), and high immune cell infiltration demonstrated significant associations with cluster 3 patients. Furthermore, five lncRNAs, namely MIR4435-2HG, AC078846.1, AL157392.3, AP001273.1, and RAD51-AS1, exhibited significant correlations with glycolysis across the five cancers. Except MIR4435-2HG, the lncRNAs were distributed in nuclei. MIR4435-2HG was connected to glycolysis, EMT, and immune infiltrations in cancers. CONCLUSIONS: We identified a subgroup of cancer patients stratified by glycolysis-associated lncRNAs with poor prognoses, high immune infiltration, and EMT activation, thus providing new directions for cancer therapy.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Glycolysis/immunology , MicroRNAs/immunology , RNA, Long Noncoding/genetics , Tumor Microenvironment/immunology , Female , Humans , Male
8.
Neurotherapeutics ; 18(2): 1371-1392, 2021 04.
Article in English | MEDLINE | ID: mdl-33410111

ABSTRACT

Limited therapeutic efficacy of temozolomide (TMZ) against glioblastomas highlights the importance of exploring new drugs for clinical therapy. Sunitinib, a multitargeted receptor tyrosine kinase inhibitor, is currently being tested as therapy for glioblastomas. Unfortunately, sunitinib still has insufficient activity to cure glioblastomas. Our aim was to determine the molecular mechanisms counteracting sunitinib drug sensitivity and find potential adjuvant drugs for glioblastoma therapy. Through in vitro experiments, transcriptome screening by RNA sequencing, and in silico analyses, we found that sunitinib induced glioma apoptotic death, and downregulated genes were enriched in oncogenic genes of glioblastoma. Meanwhile, sunitinib-upregulated genes were highly associated with the protective autophagy process. Blockade of autophagy significantly enhanced sunitinib's cytotoxicity. Growth arrest and DNA damage-inducible protein (GADD) 34 was identified as a candidate involved in sunitinib-promoted autophagy through activating p38-mitogen-activated protein kinase (MAPK) signaling. Higher GADD34 levels predicted poor survival of glioblastoma patients and induced autophagy formation in desensitizing sunitinib cytotoxicity. Guanabenz, an alpha2-selective adrenergic agonist and GADD34 functional inhibitor, was identified to enhance the efficacy of sunitinib by targeting GADD34-induced protective autophagy in glioblastoma cells, TMZ-resistant cells, hypoxic cultured cells, sphere-forming cells, and colony formation abilities. A better combined treatment effect with sunitinib and guanabenz was also observed by using xenograft mice. Taken together, the sunitinib therapy combined with guanabenz in the inhibition of GADD34-enhanced protective autophagy may provide a new therapeutic strategy for glioblastoma.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Autophagy/drug effects , Brain Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Glioblastoma/genetics , Guanabenz/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 1/antagonists & inhibitors , Sunitinib/pharmacology , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Autophagy/genetics , Brain Neoplasms/drug therapy , Cell Line, Tumor , Computer Simulation , Drug Therapy, Combination , Gene Expression Profiling , Glioblastoma/drug therapy , Humans , Mice , Molecular Targeted Therapy , Neoplasm Transplantation , Protein Phosphatase 1/genetics , RNA-Seq , Temozolomide/therapeutic use , Up-Regulation
9.
Taiwan J Obstet Gynecol ; 59(5): 698-705, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32917321

ABSTRACT

OBJECTIVE: The aim of this study was to examine the antitumor activity of hinokitiol for its clinical application in the treatment of human cervical carcinoma. MATERIALS AND METHODS: Cervical carcinoma HeLa cells were treated by different concentrations of hinokitiol. Flow cytometry was used to analyze cell cycle. Senescence-associated ß-galactosidase (SA-ß-gal) assay was used to identify senescent cells. The effects of hinokitiol on EGF-induced cell migration were determined by wound healing and transwell migration assays. Western blot was used to detect proteins involved in cell cycle progression, apoptosis, autophagy, and EGF-induced signaling pathways. RESULTS: Hinokitiol suppressed cell viability in a dose-dependent manner. Flow cytometric analysis indicated that hinokitiol treatment resulted in cell cycle arrest at G1 phase, with reduced number of cells in the G2/M phase. Western blot analysis further demonstrated that hinokitiol treatment increased the levels of p53 and p21, and concomitantly reduced the expression of cell cycle regulatory proteins, including cyclin D and cyclin E. SA-ß-gal assay showed that hinokitiol treatment significantly induced ß-galactosidase activity. In addition, treatment with hinokitiol increased the accumulation of the autophagy regulators, beclin 1 and microtubule-associated protein 1 light chain 3 (LC3-II), in a dose-dependent manner; however, it did not induce caspase-3 activation and poly ADP ribose polymerase (PARP) cleavage. In addition, epidermal growth factor-induced cell migration and c-Jun N-terminal kinase (JNK) and focal adhesion kinase (FAK) phosphorylation were significantly inhibited by hinokitiol. CONCLUSION: Our findings revealed that hinokitiol might serve as a potential therapeutic agent for cervical carcinoma therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacokinetics , Apoptosis/drug effects , Cell Movement/drug effects , Monoterpenes/pharmacology , Tropolone/analogs & derivatives , Uterine Cervical Neoplasms/drug therapy , Epidermal Growth Factor , Female , HeLa Cells , Humans , Tropolone/pharmacology
10.
Life Sci ; 254: 117807, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32422304

ABSTRACT

AIMS: Xanthohumol (XN), a natural prenylated flavonoid isolated from Humulus lupulus L. (hops), possess the therapeutic effects in glioblastoma multiforme (GBM), which is a grade IV aggressive glioma in adults. However, low bioavailability and extractive yield limit the clinical applications of XN. To comprehensively investigate XN-mediated gene networks in inducing cell death is helpful for drug development and cancer research. Therefore, we aim to identify the detailed molecular mechanisms of XN's effects on exhibiting cytotoxicity for GBM therapy. METHODS AND KEY FINDINGS: XN significantly induced GBM cell death and enhanced temozolomide (TMZ) cytotoxicity, a first-line therapeutic drug of GBM. XN-mediated transcriptome profiles and canonical pathways were identified. DNA repair signaling, a well-established mechanism against TMZ cytotoxicity, was significantly correlated with XN-downregulated genes. Replication factor C subunit 2 (RFC2), a DNA repair-related gene, was obviously downregulated in XN-treated cells. Higher RFC2 levels which occupied poor patient survival were also observed in high grade GBM patients and tumors. Inhibition of RFC2 reduced cell viability, induced cell apoptosis, and enhanced both XN and TMZ cytotoxicity. By intersecting array data, bioinformatic prediction, and in vitro experiments, microRNA (miR)-4749-5p, a XN-upregulated microRNA, was identified to target to RFC2 3'UTR and inhibited RFC2 expression. A negative correlation existed between miR-4749-5p and RFC2 in GBM patients. Overexpression of miR-4749-5p significantly promoted XN- and TMZ-mediated cytotoxicity, and reduced RFC2 levels. SIGNIFICANCE: Consequently, we suggest that miR-4749-5p targeting RFC2 signaling participates in XN-enhanced TMZ cytotoxicity of GBM. Our findings provide new potential therapeutic directions for future GBM therapy.


Subject(s)
Cell Survival/drug effects , Flavonoids/pharmacology , Glioblastoma/physiopathology , MicroRNAs/physiology , Propiophenones/pharmacology , Replication Protein C/biosynthesis , Temozolomide/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Replication Protein C/antagonists & inhibitors , Signal Transduction
11.
Neurotherapeutics ; 17(3): 1212-1227, 2020 07.
Article in English | MEDLINE | ID: mdl-31916238

ABSTRACT

DNA damage-inducible transcript 4 (DDIT4) is known to participate in various cancers, including glioblastoma multiforme (GBM). However, contradictory roles of DDIT4 exist in inducing cell death and possessing anti-apoptotic functions against cancer progression. Herein, we investigated DDIT4 signaling in GBM and temozolomide (TMZ) drug resistance. We identified that TMZ induced DDIT4 upregulation, leading to desensitization against TMZ cytotoxicity in GBM cells. Higher DDIT4 levels were found in glioma cells and mesenchymal-type GBM patients, and these higher levels were positively correlated with mesenchymal markers. Furthermore, patients with lower DDIT4 levels, especially O-6-methylguanine-DNA methyltransferase (MGMT)-methylated patients, exhibited better TMZ therapeutic efficacy. We determined that higher levels of 5 DDIT4-associated downstream genes, including SLC2A3 (also known as glucose transporter 3 (GLUT3)), can be used to predict a poor prognosis. Among these 5 genes, only GLUT3 was upregulated in both TMZ-treated and DDIT4-overexpressing cells. DDIT4-mediated GLUT3 expression was also identified, and its expression decreased TMZ's cytotoxicity. A significant correlation existed between DDIT4 and GLUT3. DDIT4 signaling was found to be involved in both glycolytic and autophagic pathways. However, GLUT3 only participated in the exhibition of DDIT4-mediated stemness, resulting from glycolytic regulation, but not in DDIT4-mediated autophagic signaling. Finally, we identified TMZ-upregulated activating transcription factor 4 (ATF4) as an upstream regulator of DDIT4-mediated GLUT3/stemness signaling and autophagy. Consequently, ATF4/DDIT4 signaling was connected to both autophagy and GLUT3-regulated stemness, which are involved in TMZ drug resistance and the poor prognoses of GBM patients. Targeting DDIT4/GLUT3 signaling might be a new direction for glioma therapy.


Subject(s)
Brain Neoplasms/metabolism , DNA Damage/physiology , Glioblastoma/metabolism , Glucose Transporter Type 3/biosynthesis , Temozolomide/therapeutic use , Transcription Factors/biosynthesis , Adolescent , Adult , Aged , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Autophagy/drug effects , Autophagy/physiology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Child , DNA Damage/drug effects , Glioblastoma/drug therapy , Glioblastoma/genetics , Glucose Transporter Type 3/genetics , Humans , Infant , Middle Aged , Temozolomide/pharmacology , Transcription Factors/genetics , Treatment Outcome
12.
PLoS One ; 14(12): e0225913, 2019.
Article in English | MEDLINE | ID: mdl-31805126

ABSTRACT

Temozolomide (TMZ) is a first-line alkylating agent for glioblastoma multiforme (GBM). Clarifying the mechanisms inducing TMZ insensitivity may be helpful in improving its therapeutic effectiveness against GBM. Insulin-like growth factor (IGF)-1 signaling and micro (mi)RNAs are relevant in mediating GBM progression. However, their roles in desensitizing GBM cells to TMZ are still unclear. We aimed to identify IGF-1-mediated miRNA regulatory networks that elicit TMZ insensitivity for GBM. IGF-1 treatment attenuated TMZ cytotoxicity via WNT/ß-catenin signaling, but did not influence glioma cell growth. By miRNA array analyses, 93 upregulated and 148 downregulated miRNAs were identified in IGF-1-treated glioma cells. miR-513a-5p from the miR-513a-2 gene locus was upregulated by IGF-1-mediated phosphoinositide 3-kinase (PI3K) signaling. Its elevated levels were also observed in gliomas versus normal cells, in array data of The Cancer Genome Atlas (TCGA), and the GSE61710, GSE37366, and GSE41032 datasets. In addition, lower levels of neural precursor cell-expressed developmentally downregulated 4-like (NEDD4L), an E3 ubiquitin protein ligase that inhibits WNT signaling, were found in gliomas by analyzing cells, arrays, and RNA sequencing data of TCGA glioma patients. Furthermore, a negative correlation was identified between miR-513a-5p and NEDD4L in glioma. NEDD4L was also validated as a direct target gene of miR-513a-5p, and it was reduced by IGF-1 treatment. Overexpression of NEDD4L inhibited glioma cell viability and reversed IGF-1-repressed TMZ cytotoxicity. In contrast, miR-513a-5p significantly affected NEDD4L-inhibited WNT signaling and reduced TMZ cytotoxicity. These findings demonstrate a distinct role of IGF-1 signaling through miR-513a-5p-inhibited NEDD4L networks in influencing GBM's drug sensitivity to TMZ.


Subject(s)
Drug Resistance, Neoplasm , Glioma/genetics , Glioma/metabolism , Insulin-Like Growth Factor I/metabolism , MicroRNAs/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Temozolomide/pharmacology , Wnt Signaling Pathway/drug effects , 3' Untranslated Regions , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , RNA Interference
13.
Pharmacol Res ; 147: 104390, 2019 09.
Article in English | MEDLINE | ID: mdl-31398406

ABSTRACT

Temozolomide (TMZ) is a first-line chemotherapeutic agent used against glioblastoma multiforme (GBM), but this disease exhibits recurrence and high lethality. Therefore, it is critical to explore biomarkers which involve in drug resistance and can be represented as different therapeutic effects after a diagnosis. We attempted to investigate the underlying variably expressed genes that contribute to the formation of resistance to TMZ. We analyzed gene and microRNA (miR) data from GBM patients in The Cancer Genome Atlas (TCGA) database to identify genetic factors associated with poor TMZ efficacy. By conducting a gene set enrichment analysis (GSEA), the epithelial-to-mesenchymal transition (EMT) was associated with poor TMZ responses. To identify roles of microRNAs in regulating TMZ resistance, a differential microRNA analysis was performed in TMZ-treated GBM patients. Downregulation of miR-140 was significantly correlated with poor survival. By integrating TCGA transcriptomic data and genomics of drug sensitivity in cancer (GDSC), cathepsin B (CTSB) was inversely associated with miR-140 expression and poor TMZ efficacy. By a pan-cancer analysis, both miR-140 and CTSB were found to be prognostic factors in other cancer types. We also identified that CTSB was a direct target gene of miR-140. Overexpression of miR-140 reduced CTSB levels, enhanced TMZ cytotoxicity, suppressed the mesenchymal transition, and influenced CTSB-regulated tumor sphere formation and stemness marker expression. In contrast, overexpression of CTSB decreased TMZ-induced glioma cell death, promoted the mesenchymal transition, and attenuated miR-140-increased TMZ cytotoxicity. These findings provide novel targets to increase the therapeutic efficacy of TMZ against GBM.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Cathepsin B/genetics , Epithelial-Mesenchymal Transition , Glioblastoma/drug therapy , MicroRNAs , Temozolomide/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Proportional Hazards Models , RNA, Messenger/metabolism , Signal Transduction/drug effects , Temozolomide/therapeutic use
14.
PLoS One ; 13(12): e0208459, 2018.
Article in English | MEDLINE | ID: mdl-30521597

ABSTRACT

Tumor-infiltrating lymphocytes are related to positive clinical prognoses in numerous cancer types. Programmed death ligand 1 (PD-L1), a mediator of the PD-1 receptor, plays an inhibitory role in cancer immune responses. PD-L1 upregulation can impede infiltrating T-cell functions in lung adenocarcinoma (LUAD), a lung cancer subtype. However, associations between the expression of PD-L1 and infiltration of B cells (a major immunoregulatory cell) remain unknown. Therefore, we investigated the role of infiltrating B cells in LUAD progression and its correlation with PD-L1 expression. The Cancer Genome Atlas (TCGA) LUAD data set was used to explore associations among B-cell infiltration, PD-L1 expression, clinical outcome, and gene landscape. Gene set enrichment analysis was used to explore putative signaling pathways and candidate genes. The drug enrichment analysis was used to identify candidate genes and the related drugs. We found that high B-cell infiltration was correlated with better prognoses; however, PD-L1 may interfere with the survival advantage in patients with high B-cell infiltration. The gene landscape was characterized comprehensively, with distinct PD-L1 levels in cell populations with high B-cell infiltration. We obtained five upregulated signaling pathways from the gene landscape: apoptosis, tumor necrosis factor (TNF)-α signaling via nuclear factor (NF)-κB, apical surface, interferon-α response, and KRAS signaling. Moreover, four candidate genes and their related target drugs were also identified, namely interleukin-2ß receptor (IL2RB), IL-2γ receptor (IL2RG), Toll-like receptor 8 (TLR8), and TNF. These findings suggest that tumor-infiltrating B cells could act as a clinical factor in anti-PD-L1 immunotherapy for LUAD.


Subject(s)
Adenocarcinoma of Lung/drug therapy , B-Lymphocytes/metabolism , B7-H1 Antigen/genetics , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Aged , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Middle Aged , Prognosis , Sequence Analysis, RNA , Signal Transduction , Survival Analysis , Up-Regulation
15.
Chem Biol Interact ; 291: 144-151, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29890129

ABSTRACT

Imatinib (IM) is a first-line therapeutic drug for chronic myeloid leukemia (CML), a hematological disease. Mutations in the BCR-ABL domain increase formation of IM resistance in CML. However, not all patients are BCR-ABL domain-mutant dependent. Investigating non-mutant mechanisms in the development of acquired IM resistance is a critical issue. We explored the mechanisms which influence IM efficacy and resistance in CML. Higher protective autophagy was identified in IM-resistant K562 (K562R) cells. Inhibition of autophagy by the inhibitors, chloroquine and 3-methyladenine, enhanced IM's efficacy in K562R cells. In addition, microRNA (miR)-199a/b-5p were downregulated in K562R cells compared to parent cells. Overexpression of miR-199a/b-5p reduced autophagy and induced cell apoptosis, resulting in enhanced IM's efficacy in K562R cells. Moreover, expression levels of the Wingless-type MMTV integration site family member 2 (WNT2), a positive regulator of autophagy, were significantly higher in K562R cells, and it was validated as a direct target gene of miR-199a/b-5p. Overexpressions of miR-199a/b-5p inhibited WNT2 downstream signaling. Furthermore, overexpression and knockdown of WNT2 influenced autophagy formation and CML drug sensitivity to IM. Overexpression of WNT2 could also reverse miR-199a/b-5p-enhanced IM efficacy in K562R cells. These results emphasized that miR-199a/b-5p inhibited autophagy via repressing WNT2 signaling and might provide novel therapeutic strategies for future IM-resistant CML therapy and drug development.


Subject(s)
Autophagy , Drug Resistance, Neoplasm , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/metabolism , Signal Transduction , Wnt2 Protein/metabolism , 3' Untranslated Regions/genetics , Apoptosis/drug effects , Autophagy/drug effects , Base Sequence , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Imatinib Mesylate/pharmacology , Signal Transduction/drug effects , Treatment Outcome
16.
J Neurochem ; 146(3): 269-288, 2018 08.
Article in English | MEDLINE | ID: mdl-29747239

ABSTRACT

Glioblastoma multiforme is the most common brain tumor in adults. Because of its highly invasive nature, it is not easy to treat, resulting in high mortality rates. Stromal interacting molecule 1 (Stim1) plays important roles in regulating store-operated Ca2+ entry, and controls invasion by cancer cells. However, the mechanisms and functions of Stim1 in glioma progression are still unclear. In this study, we investigated the effects of targeting Stim1 expression on glioma cell invasion. By analyzing profiles of glioblastoma multiforme patients from RNA-sequencing data in The Cancer Genome Atlas, higher expression levels of STIM1 were correlated with the poor survival. Furthermore, signaling pathways associated with tumor malignancy, including the epithelial-to-mesenchymal transition (EMT), were activated in patients with high STIM1 expression according to gene set enrichment analyses. Higher Stim1 levels were found in glioma cells compared to human astrocytes, and these higher levels enhanced glioma cell invasion. Xanthohumol (XN), a prenylated flavonoid extracted from the hop plant Humulus lupulus L. (Cannabaceae), significantly reduced cell invasion through inhibiting Stim1 expression. From an micro(mi)RNA array analysis, miR-4725-3p was up-regulated by XN treatment. Over-expression of miR-4725-3p inhibited glioma cell invasion via directly targeting the 3'-untranslated region of STIM1. The extracellular signal-regulated kinase/c-Fos pathway was also validated to participate in XN-up-regulated miR-4725-3p expression according to promoter and chromatin immunoprecipitation assays. These results emphasize that miR-4725-3p-inhibited STIM1 signaling is involved in XN-attenuated glioma cell invasion. These findings may provide insights into novel therapeutic strategies for future glioblastoma therapy and drug development. Open Data: Materials are available on https://cos.io/our-services/open-science-badges/ https://osf.io/93n6m/.


Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Propiophenones/pharmacology , Signal Transduction/drug effects , Stromal Interaction Molecule 1/metabolism , Aged , Aged, 80 and over , Cell Line, Tumor , Chromatin Immunoprecipitation , Collagen , Drug Combinations , Female , Humans , Laminin , Male , MicroRNAs/genetics , Middle Aged , Mutagenesis/drug effects , Mutagenesis/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Proteoglycans , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Stromal Interaction Molecule 1/genetics
17.
Sci Rep ; 7(1): 732, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28389653

ABSTRACT

The insulin-like growth factor (IGF)-1 signaling is relevant in regulating cell growth and cytokine secretions by glioblastomas. MicroRNAs determine the cell fate in glioblastomas. However, relationships between IGF-1 signaling and miRNAs in glioblastoma pathogenesis are still unclear. Our aim was to validate the IGF-1-mediated mRNA/miRNA regulatory network in glioblastomas. Using in silico analyses of mRNA array and RNA sequencing data from The Cancer Genome Atlas (TCGA), we identified 32 core enrichment genes that were highly associated with IGF-1-promoted cytokine-cytokine receptor interactions. To investigate the IGF-1-downregulated miRNA signature, microarray-based approaches with IGF-1-treated U87-MG cells and array data in TCGA were used. Four miRNAs, including microRNA (miR)-9-5p, miR-9-3p, miR-181d, and miR-130b, exhibited an inverse correlation with IGF-1 levels. The miR-181d, that targeted the most IGF-1-related cytokine genes, was significantly reduced in IGF-1-treated glioma cells. Statistical models incorporating both high-IGF-1 and low-miR-181d statuses better predicted poor patient survival, and can be used as an independent prognostic factor in glioblastomas. The C-C chemokine receptor type 1 (CCR1) and interleukin (IL)-1b demonstrated inverse correlations with miR-181d levels and associations with patient survival. miR-181d significantly attenuated IGF-1-upregulated CCR1 and IL-1b gene expressions. These findings demonstrate a distinct role for IGF-1 signaling in glioma progression via miR-181d/cytokine networks.


Subject(s)
Cytokines/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Insulin-Like Growth Factor I/genetics , MicroRNAs/genetics , RNA Interference , RNA, Messenger/genetics , 3' Untranslated Regions , Cell Line, Tumor , Computational Biology/methods , Gene Expression Profiling , Gene Regulatory Networks , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Insulin-Like Growth Factor I/metabolism , Prognosis , Transcriptome
18.
PLoS One ; 12(3): e0173890, 2017.
Article in English | MEDLINE | ID: mdl-28323865

ABSTRACT

MicroRNAs are small noncoding RNAs that post-transcriptionally control the expression of genes involved in glioblastoma multiforme (GBM) development. Although miR-302b functions as a tumor suppressor, its role in GBM is still unclear. Therefore, this study comprehensively explored the roles of miR-302b-mediated gene networks in GBM cell death. We found that miR-302b levels were significantly higher in primary astrocytes than in GBM cell lines. miR-302b overexpression dose dependently reduced U87-MG cell viability and induced apoptosis through caspase-3 activation and poly(ADP ribose) polymerase degradation. A transcriptome microarray revealed 150 downregulated genes and 380 upregulated genes in miR-302b-overexpressing cells. Nuclear factor IA (NFIA), higher levels of which were significantly related to poor survival, was identified as a direct target gene of miR-302b and was involved in miR-302b-induced glioma cell death. Higher NFIA levels were observed in GBM cell lines and human tumor sections compared with astrocytes and non-tumor tissues, respectively. NFIA knockdown significantly enhanced apoptosis. We found high levels of insulin-like growth factor-binding protein 2 (IGFBP2), another miR-302b-downregulated gene, in patients with poor survival. We verified that NFIA binds to the IGFBP2 promoter and transcriptionally enhances IGFBP2 expression levels. We identified that NFIA-mediated IGFBP2 signaling pathways are involved in miR-302b-induced glioma cell death. The identification of a regulatory loop whereby miR-302b inhibits NFIA, leading to a decrease in expression of IGFBP-2, may provide novel directions for developing therapies to target glioblastoma tumorigenesis.


Subject(s)
Glioma/genetics , Glioma/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , NFI Transcription Factors/antagonists & inhibitors , NFI Transcription Factors/genetics , Apoptosis/genetics , Apoptosis/physiology , Astrocytes/cytology , Astrocytes/metabolism , Cell Line, Tumor , Cell Survival/genetics , Down-Regulation , Gene Knockdown Techniques , Gene Regulatory Networks , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/pathology , Humans , Insulin-Like Growth Factor Binding Protein 2/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 2/metabolism , NFI Transcription Factors/metabolism , Promoter Regions, Genetic , Signal Transduction , Transcriptome
19.
Neuropharmacology ; 116: 300-314, 2017 04.
Article in English | MEDLINE | ID: mdl-27986595

ABSTRACT

Glioblastoma multiforme (GBM) is the high-grade primary glioma in adults. Temozolomide (TMZ), an alkylating agent of the imidazotetrazine series, is a first-line chemotherapeutic drug for clinical therapy. However, the expense of TMZ therapy and increasing drug resistance to TMZ decreases its therapeutic effects. Therefore, our aim was to investigate the detailed molecular mechanisms of TMZ-mediated cytotoxicity to enhance the efficacy of TMZ in clinical GBM therapy. First, TMZ-mediated gene expression profiles and networks in U87-MG cells were identified by transcriptome microarray and bioinformatic analyses. Cation transport regulator-like protein 1 (CHAC1) was the most highly TMZ-upregulated gene. Overexpression and knockdown of CHAC1 expression significantly influenced TMZ-mediated cell viability, apoptosis, caspase-3 activation, and poly(ADP ribose) polymerase (PARP) degradation. The c-Jun N-terminal kinase (JNK)1/c-JUN pathway was identified to participate in TMZ-upregulated CHAC1 expression via transcriptional control. Furthermore, CHAC1 levels were significantly decreased in GBM cell lines, TCGA array data, and tumor tissues. Overexpression of CHAC1 enhanced glioma apoptotic death via caspase-3/9 activation, PARP degradation, autophagy formation, reactive oxygen species generation, increased intracellular calcium, and loss of the mitochondria membrane potential. Finally, we also identified that TMZ significantly reduced Notch3 levels, which are upregulated in gliomas. TMZ also induced CHAC1 to bind to the Notch3 protein and inhibit Notch3 activation, resulting in attenuation of Notch3-mediated downstream signaling pathways. These results emphasize that CHAC1-inhibited Notch3 signaling can influence TMZ-mediated cytotoxicity. Our findings may provide novel therapeutic strategies for future glioblastoma therapy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Receptor, Notch3/metabolism , gamma-Glutamylcyclotransferase/pharmacology , Antineoplastic Agents, Alkylating/toxicity , Autophagy/drug effects , Autophagy/physiology , Calcium/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dacarbazine/pharmacology , Dacarbazine/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Glioma/metabolism , Glioma/pathology , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitogen-Activated Protein Kinase 8/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Temozolomide , gamma-Glutamylcyclotransferase/toxicity
20.
PLoS One ; 11(11): e0167096, 2016.
Article in English | MEDLINE | ID: mdl-27893811

ABSTRACT

Temozolomide (TMZ), an alkylating agent of the imidazotetrazine series, is a first-line chemotherapeutic drug used in the clinical therapy of glioblastoma multiforme, the most common and high-grade primary glioma in adults. Micro (mi)RNAs, which are small noncoding RNAs, post-transcriptionally regulate gene expressions and are involved in gliomagenesis. However, no studies have reported relationships between TMZ and miRNA gene regulation. We investigated TMZ-mediated miRNA profiles and its molecular mechanisms underlying the induction of glioma cell death. By performing miRNA microarray and bioinformatics analyses, we observed that expression of 248 miRNAs was altered, including five significantly upregulated and 17 significantly downregulated miRNAs, in TMZ-treated U87MG cells. miR-128 expression levels were lower in different glioma cells and strongly associated with poor survival. TMZ treatment significantly upregulated miR-128 expression. TMZ significantly enhanced miR-128-1 promoter activity and transcriptionally regulated miR-128 levels through c-Jun N-terminal kinase 2/c-Jun pathways. The overexpression and knockdown of miR-128 expression significantly affected TMZ-mediated cell viability and apoptosis-related protein expression. Furthermore, the overexpression of miR-128 alone enhanced apoptotic death of glioma cells through caspase-3/9 activation, poly(ADP ribose) polymerase degradation, reactive oxygen species generation, mitochondrial membrane potential loss, and non-protective autophagy formation. Finally, we identified that key members in mammalian target of rapamycin (mTOR) signaling including mTOR, rapamycin-insensitive companion of mTOR, insulin-like growth factor 1, and PIK3R1, but not PDK1, were direct target genes of miR-128. TMZ inhibited mTOR signaling through miR-128 regulation. These results indicate that miR-128-inhibited mTOR signaling is involved in TMZ-mediated cytotoxicity. Our findings may provide a better understanding of cytotoxic mechanisms of TMZ involved in glioblastoma development.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Glioma/pathology , Insulin-Like Growth Factor I/metabolism , MicroRNAs/genetics , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Dacarbazine/pharmacology , Glioma/drug therapy , Glioma/genetics , Humans , Insulin-Like Growth Factor I/genetics , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Survival Rate , TOR Serine-Threonine Kinases/genetics , Temozolomide , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...