Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Oncol ; 63(3)2023 Sep.
Article in English | MEDLINE | ID: mdl-37539753

ABSTRACT

Following the publication of this paper, and subsequently to a corrigendum that was published to take account of errors that had been made concerning the Transwell invasion assay data in Fig. 4B (doi: 10.3892/ijo.2022.5463), it was drawn to the Editor's attention by a concerned reader that the western blots shown in Figs. 3C and 6B shared the same control data; moreover, potential anomalies were identified in the flow cytometric plots shown in Fig. 2A. Although the authors proposed to publish a corrigendum to account for these errors, including new data after having performed certain of the experiments again, given the number of problems that have been identified concerning the assembly of various of the figures in the published paper, the Editor of International Journal of Oncology has decided not to publish a further corrigendum, and has, in fact, determined that this paper should be retracted from the Journal on account of an overall lack of confidence in the originally presented data. The Editor apologizes to the readership for any inconvenience caused. [International Journal of Oncology 50: 893-902, 2017; DOI: 10.3892/ijo.2017.3871].

2.
Int J Oncol ; 62(1)2023 Jan.
Article in English | MEDLINE | ID: mdl-36484397

ABSTRACT

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that Fig. 4B on p. 899, showing the results of Transwell invasion assay experiments, contained a pair of apparently overlapping panels, such that they may have been derived from the same original source, even though they were intended to show the results from differently performed experiments. After having re­examined their original data, the authors were able to identify that Fig. 4B had been inadvertently assembled incorrectly. The revised version of Fig. 4, featuring the correct data for the SB­T­121205, 10 nM data panel (the lower­left panel in Fig. 4B), is shown on the next page. The authors confirm that these data continue to support the main conclusions presented in their paper, and are grateful to the Editor of International Journal of Oncology for allowing them this opportunity to publish a Corrigendum. They also apologize to the readership for any inconvenience caused. [International Journal of Oncology 50: 893­902, 2017; DOI: 10.3892/ijo.2017.3871].

3.
Pak J Pharm Sci ; 33(1): 161-168, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32122844

ABSTRACT

Tigecycline (TGC) and cefoperazone/sulbactam (CPS) both have been shown good in vitro activity against carbapenem-resistant Acinetobacter baumannii (CRAB) isolates. We aim to compare the efficacy of TGC versus CPS for CRAB infections. We conducted a retrospective cohort study of patients with CRAB at a single center in China from 2013 to 2015. Outcomes comprised in-hospital mortality, clinical and microbiological response. The method of inverse probability of treatment weighting and multivariable logistic regression analysis incorporated with propensity score were employed to estimate the effect of treatment groups. There were 130 subjects included in our study. The patients in TGC, CPS and TGC plus CPS combination group were 42, 66, and 22, respectively. After adjustment, in-hospital mortality was lower in CPS group than TGC group (weighted OR 0.173; 95% CI 0.06-0.497; P=0.001) but without differences in clinical success and microbiological eradication (P>0.05). TGC monotherapy had a similar outcome with TGC plus CPS combination group. This is the first study comparing the efficacy of tigecycline and cefoperazone/sulbactam for CRAB infections. Cefoperazone/sulbactam appears to be more efficacious than tigecycline during treatment.


Subject(s)
Acinetobacter Infections/drug therapy , Cefoperazone/therapeutic use , Sulbactam/therapeutic use , Tigecycline/therapeutic use , Acinetobacter Infections/mortality , Acinetobacter baumannii/drug effects , Aged , Aged, 80 and over , Anti-Bacterial Agents/pharmacology , Cefoperazone/pharmacology , Drug Resistance, Bacterial , Drug Therapy, Combination/mortality , Female , Humans , Male , Middle Aged , Pentostatin/analogs & derivatives , Retrospective Studies , Sulbactam/pharmacology , Tigecycline/pharmacology
4.
Mol Cancer Ther ; 18(12): 2457-2468, 2019 12.
Article in English | MEDLINE | ID: mdl-31488699

ABSTRACT

MDR and tumor migration and invasion are still the main obstacles to effective breast cancer chemotherapies. Transgelin 2 has recently been shown to induce drug resistance, tumor migration, and invasion. The aim of this study was to determine the biological functions of Transgelin 2 and the mechanism underlying how Transgelin 2 induces paclitaxel (PTX) resistance and the migration and invasion of breast cancer. We detected that the protein level of Transgelin 2 was significantly upregulated in breast cancer tissues compared with adjacent nontumor tissues. A bioinformatics analysis indicated that Transgelin 2 was significantly related to clinicopathologic parameters and patient prognosis. Overexpression of Transgelin 2 enhanced the migration and invasion of human breast cancer cells and decreased the sensitivity of breast cancer cells to paclitaxel. Meanwhile, the tumorigenesis and metastasis of breast cancer cells were also enhanced by Transgelin 2 overexpression in vivo Moreover, Transgelin 2 overexpression activated the PI3K/Akt/GSK-3ß pathway by increasing the phosphorylation levels of Akt and GSK-3ß and decreasing the expression of PTEN. We also found that Transgelin 2 could directly interact with PTEN and was located upstream of PTEN. Furthermore, the PI3K/Akt pathway inhibitor MK-2206 reversed the resistance to paclitaxel and inhibited the migration and invasion of breast cancer cells. These findings indicate that Transgelin 2 promotes paclitaxel resistance and the migration and invasion of breast cancer by directly interacting with PTEN and activating the PI3K/Akt/GSK-3ß pathway. Transgelin 2 may therefore be useful as a novel biomarker and therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Glycogen Synthase Kinase 3 beta/metabolism , Microfilament Proteins/therapeutic use , Muscle Proteins/therapeutic use , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Female , Humans , Mice , Mice, Nude , Microfilament Proteins/pharmacology , Muscle Proteins/pharmacology
5.
J Transl Med ; 16(1): 157, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880050

ABSTRACT

BACKGROUND: Invasive pulmonary aspergillosis (IPA) is a life-threatening disease in immunosuppressed patients. Voriconazole is commonly used to prevent and treat IPA in the clinic, but the optimal prophylactic antifungal regimen is unknown. The objective of this study was to clarify the mechanism underlying how voriconazole prevents IPA based on a target cellular pharmacokinetics/pharmacodynamics model, with the aim of identifying a way to design an optimal prophylactic antifungal regimen. METHODS: A nystatin assay was used to establish a target-cells model for A. fumigatus infection. An inhibitory effect sigmoid Emax model was developed to explore the cellular PK/PD breakpoint, and Monte Carlo simulation was used to design the prophylactic antifungal regimen. RESULTS: The intracellular activity of voriconazole in the target cells varied with its concentration, with the minimum inhibitory concentration (MIC) being an important determinant. For A. fumigatus strains AF293 and AF26, voriconazole decreased the intracellular inoculum by 0.79 and 0.84 lg cfu, respectively. The inhibitory effect sigmoid Emax model showed that 84.01% of the intracellular inoculum was suppressed by voriconazole within 24 h, and that a PK/PD value of 35.53 for the extracellular voriconazole concentration divided by MIC was associated with a 50% suppression of intracellular A. fumigatus. The Monte Carlo simulation results showed that the oral administration of at least 200 mg of voriconazole twice daily was yielded estimated the cumulative fraction of response value of 91.48%. Concentration of voriconazole in the pulmonary epithelial lining fluid and the plasma of > 17.77 and > 1.55 mg/L, respectively, would ensure the PK/PD > 35.53 for voriconazole against most isolates of A. fumigatus and may will be benefit to prevent IPA in clinical applications. CONCLUSIONS: This study used a target cellular pharmacokinetics/pharmacodynamics model to reveal a potential mechanism underlying how voriconazole prevents IPA and has provided a method for designing voriconazole prophylactic antifungal regimen in immunosuppressed patients.


Subject(s)
Invasive Pulmonary Aspergillosis/drug therapy , Invasive Pulmonary Aspergillosis/prevention & control , Voriconazole/pharmacokinetics , Voriconazole/therapeutic use , A549 Cells , Aspergillus fumigatus/drug effects , Biomarkers/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Galactose/analogs & derivatives , Humans , Invasive Pulmonary Aspergillosis/microbiology , Mannans/metabolism , Microbial Sensitivity Tests , Monte Carlo Method , Regression Analysis , Voriconazole/pharmacology
6.
Acta Pharmacol Sin ; 39(9): 1533-1543, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29849129

ABSTRACT

Caspofungin is an echinocandin antifungal agent licensed as a first-line therapy for invasive candidiasis in patients with moderate to severe illness or recent exposure to azoles. In this study we developed a whole-body physiology-based pharmacokinetics (WB-PBPK) model to predict the pharmacokinetics (PK) of caspofungin, and combined with Monte Carlo simulation (MCS) to optimize clinical dosage regimens of caspofungin in different kinds of patients. A WB-PBPK model of caspofungin was built and validated with raw data from 4 previous trials of general patients, intensive care unit (ICU) patients with Child-Pugh B, ICU patients on continuous renal replacement therapy, mild and moderate hepatic insuffciency (HI) patients. MCS was used to optimize clinical dosage regimens of caspofungin in these patients. A cumulative fraction of response (CFR) value of ≥90% was considered to be the minimum for achieving optimal empirical therapy. The simulated results of the WB-PBPK model were in good agreement with observed values of all trials. For general and ICU patients with caspofungin 70/50 mg, AUC and Cmax were decreased with the increase of body weight (BW) and showed great variation. MCS showed all general patients achieved CFR≥90% regardless of BW. But not all ICU patients with higher BW (≥70 kg) could achieve CFR≥90%. Compared with standard dosage regimens in general patients, caspofungin 70/35 mg in ICU patients with Child-Pugh B achieved significantly decreased AUC and Cmax, but obtained similar AUC and Cmax in moderate HI patients with Child-Pugh B. The WB-PBPK model of caspofungin is able to predict PK of all populations correctly. The combined WB-PBPK model with MCS can successfully optimize clinical dosage regimens of caspofungin in all patient populations.


Subject(s)
Antifungal Agents/pharmacokinetics , Caspofungin/pharmacokinetics , Models, Biological , Adult , Antifungal Agents/administration & dosage , Caspofungin/administration & dosage , Hepatic Insufficiency/metabolism , Humans , Intensive Care Units , Male , Monte Carlo Method , Young Adult
7.
BMC Infect Dis ; 18(1): 155, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29609553

ABSTRACT

BACKGROUND: Posaconazole therapeutic drug monitoring (TDM) is increasingly used in clinical practice. However, the utility of posaconazole TDM and the target of posaconazole plasma concentration for clinical successful prophylaxis remain uncertain and controversial. The aim of this study was to evaluate posaconazole exposure-response relationship and determine an optimum posaconazole concentration for prophylaxis against invasive fungal infections (IFIs). METHODS: Bibliographic databases were searched (from inception to September 2017) to select studies including the clinical outcomes below and above concentration cut-off value of 0.5 mg/L and 0.7 mg/L. The reliability of the results were evaluated with trial sequential analysis (TSA). RESULTS: Twenty-eight studies with 1930 patients included were analyzed. The results of our pooled analysis demonstrated that patients with posaconazole plasma concentrations over 0.5 mg/L were twice more likely to achieve successful responses compared with those with lower concentrations (odds ratio, OR = 1.98, 95% confidence interval, CI 1.09-3.58, P = 0.02) while the threshold, 0.7 mg/L showed no significant difference (OR = 1.84, 95% CI 0.94-3.63, P = 0.08). The TSA results showed that there was sufficient information to support these findings. CONCLUSIONS: An optimal posaconazole concentration target of 0.5 mg/L is suggested to ensure the clinical prophylactic efficacy and may help reduce the dosage and dose-dependent toxicity comparing with the target of 0.7 mg/L.


Subject(s)
Antifungal Agents/therapeutic use , Drug Monitoring/methods , Invasive Fungal Infections/drug therapy , Mycoses/drug therapy , Triazoles/therapeutic use , Antifungal Agents/blood , Blood Chemical Analysis , Chemoprevention/methods , Humans , Invasive Fungal Infections/blood , Mycoses/blood , Predictive Value of Tests , Reproducibility of Results , Treatment Outcome , Triazoles/blood
8.
Endocr Relat Cancer ; 25(5): 533-545, 2018 05.
Article in English | MEDLINE | ID: mdl-29555649

ABSTRACT

Thyroid hormone, l-thyroxine (T4), has been shown to promote ovarian cancer cell proliferation via a receptor on plasma membrane integrin αvß3 and to induce the activation of ERK1/2 and expression of programmed death-ligand 1 (PD-L1) in cancer cells. In contrast, resveratrol binds to integrin αvß3 at a discrete site and induces p53-dependent antiproliferation in malignant neoplastic cells. The mechanism of resveratrol action requires nuclear accumulation of inducible cyclooxygenase (COX)-2 and its complexation with phosphorylated ERK1/2. In this study, we examined the mechanism by which T4 impairs resveratrol-induced antiproliferation in human ovarian cancer cells and found that T4 inhibited resveratrol-induced nuclear accumulation of COX-2. Furthermore, T4 increased expression and cytoplasmic accumulation of PD-L1, which in turn acted to retain inducible COX-2 in the cytoplasm. Knockdown of PD-L1 by small hairpin RNA (shRNA) relieved the inhibitory effect of T4 on resveratrol-induced nuclear accumulation of COX-2- and COX-2/p53-dependent gene expression. Thus, T4 inhibits COX-2-dependent apoptosis in ovarian cancer cells by retaining inducible COX-2 with PD-L1 in the cytoplasm. These findings provide new insights into the antagonizing effect of T4 on resveratrol's anticancer properties.


Subject(s)
Apoptosis/drug effects , B7-H1 Antigen/metabolism , Ovarian Neoplasms/drug therapy , Resveratrol/metabolism , Thyroxine/therapeutic use , Female , Humans , Ovarian Neoplasms/pathology , Thyroxine/pharmacology , Transfection
9.
J Drug Target ; 26(1): 27-35, 2018 01.
Article in English | MEDLINE | ID: mdl-28562134

ABSTRACT

Multidrug resistance (MDR) is a serious obstacle in breast cancer patients which limits chemotherapeutic drugs application. Our previous study confirmed that overexpression of nucleophosmin (NPM) was closely related to MDR in methotrexate-resistant breast cancer cells (MCF-7/MTX), and NPM could be a potential therapeutic target for chemoresistance. In this work, we aim to investigate NPM-mediated resistance mechanism in breast carcinoma. The NPM level was strongly positive in breast carcinoma tissues compared with adjacent normal samples, which was associated with lymph node metastasis. We found abnormal expression of NPM activated PI3K/Akt pathway and affected downstream apoptosis factors. Then, NPM level was attenuated by RNA interfering technology, the sensitivity of MCF-7/MTX cells to methotrexate was obviously increased, factor level of mitochondria apoptosis pathway was significantly augmented, and Akt phosphorylation was inhibited. Furthermore, examination of Akt and NPM level demonstrated that Akt inhibitor MK-2206 sensitised resistant cells to methotrexate and induced MCF-7/MTX cell apoptosis by PI3K/Akt pathway and mitochondria apoptosis pathway. These suggested NPM-induced resistance and anti-apoptosis were required for Akt activity. NPM has a crucial function in MDR of breast cancer through influencing Akt activity and resistant cell apoptosis, and it could be expected to become a therapeutic target for chemoresistance in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Female , Gene Knockdown Techniques , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , MCF-7 Cells , Methotrexate/pharmacology , Middle Aged , Nuclear Proteins/genetics , Nucleophosmin , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
10.
Int J Hematol ; 107(2): 235-243, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29027638

ABSTRACT

This study evaluated the clinical and cost-effectiveness of prophylactic use of fluconazole versus mould-active triazoles (voriconazole and posaconazole) in adult patients with acute lymphoblastic leukemia (ALL). A decision analytical model was developed with inputs from a 7-year retrospective study (2009-2016) of 103 consecutive adult patients with ALL who received antifungal prophylaxis. Information on the administration of antifungal agents, clinical outcomes, and costs were collected. One-way sensitivity analyses and probabilistic sensitivity analysis were performed. The mould-active triazoles group was associated with higher life-years (3.71 vs 3.59) and lower total costs (US$4886 vs US$5722) per patient compared with fluconazole. One-way sensitivity analyses revealed that varying all of the key variables in the model did not affect the robustness of the results. Probabilistic sensitivity analysis demonstrated that mould-active triazoles had a probability of 77.1 and 90.1% of providing a dominant and cost-effective option relative to fluconazole, respectively. Mould-active triazoles should be regarded as preferable to fluconazole as the first-line prophylactic for adult patients with ALL accompanied by uncommon severe vinca alkaloid-induced neurotoxicity. However, the results reported here should be interpreted with caution owing to the observational nature of the data.


Subject(s)
Antibiotic Prophylaxis , Antifungal Agents/administration & dosage , Antifungal Agents/economics , Fluconazole/administration & dosage , Fluconazole/economics , Mycoses/complications , Mycoses/prevention & control , Opportunistic Infections/complications , Opportunistic Infections/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Triazoles/administration & dosage , Triazoles/economics , Adult , Aged , Antibiotic Prophylaxis/economics , Cost-Benefit Analysis , Female , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Young Adult
11.
Article in English | MEDLINE | ID: mdl-28993334

ABSTRACT

The use of antifungal interventions in critically ill patients prior to invasive fungal infection (IFI) being microbiologically confirmed and the preferred drug are still controversial. A systematic literature search was performed to identify randomized controlled trials (RCTs) that compared untargeted antifungal treatments applied to nonneutropenic critically ill patients. The primary outcomes were all-cause mortality and proven IFI rates. A random-effects model was used with trial sequential analyses (TSA), a network meta-analysis (NMA) was conducted to obtain indirect evidence, and a cost-effectiveness analysis using a decision-analytic model was completed from the patient perspective over a lifetime horizon. In total, 19 RCTs involving 2,556 patients (7 interventions) were included. Untargeted antifungal treatment did not significantly decrease the incidence of all-cause mortality (odds ratio [OR] = 0.89, 95% confidence interval [95%CI] = 0.70 to 1.14), but it did reduce the incidence of proven IFI (OR = 0.45, 95%CI = 0.29 to 0.71) relative to placebo/no intervention. The TSA showed that there was sufficient evidence supporting these findings. In the NMA, the only significant difference found for both primary outcomes was between fluconazole and placebo/no intervention in preventing proven IFI (OR = 0.35, 95%CI = 0.19 to 0.65). Based on drug and hospital costs in China, the incremental cost-effectiveness ratios per life-year saved for fluconazole, caspofungin, and micafungin relative to placebo/no intervention corresponded to US$889, US$9,994, and US$10,351, respectively. Untargeted antifungal treatment significantly reduced proven IFI rates in nonneutropenic critically ill patients but with no mortality benefits relative to placebo/no intervention. Among the well-tolerated antifungals, fluconazole remains the only one that is effective for IFI prevention and significantly cheaper than echinocandins.


Subject(s)
Antifungal Agents/therapeutic use , Cost-Benefit Analysis/methods , Economics, Pharmaceutical , Invasive Fungal Infections/prevention & control , Primary Prevention/economics , Primary Prevention/methods , Antifungal Agents/economics , Caspofungin , Critical Illness/mortality , Echinocandins/economics , Echinocandins/therapeutic use , Fluconazole/economics , Fluconazole/therapeutic use , Humans , Invasive Fungal Infections/drug therapy , Lipopeptides/economics , Lipopeptides/therapeutic use , Micafungin , Network Meta-Analysis
12.
Ann N Y Acad Sci ; 1403(1): 92-100, 2017 09.
Article in English | MEDLINE | ID: mdl-28759712

ABSTRACT

Nonpeptide hormones, such as thyroid hormone, dihydrotestosterone, and estrogen, have been shown to stimulate cancer proliferation via different mechanisms. Aside from their cytosolic or membrane-bound receptors, there are receptors on integrin αv ß3 for nonpeptide hormones. Interaction between hormones and integrin αv ß3 can induce signal transduction and eventually stimulate cancer cell proliferation. Resveratrol induces inducible COX-2-dependent antiproliferation via integrin αv ß3 . Resveratrol and hormone-induced signals are both transduced by activated extracellular-regulated kinases 1 and 2 (ERK1/2); however, hormones promote cell proliferation, while resveratrol induces antiproliferation in cancer cells. Hormones inhibit resveratrol-stimulated phosphorylation of p53 on Ser15, resveratrol-induced nuclear COX-2 accumulation, and formation of p53-COX-2 nuclear complexes. Subsequently, hormones impair resveratrol-induced COX-2-/p53-dependent gene expression. The inhibitory effects of hormones on resveratrol action can be blocked by different antagonists of specific nonpeptide hormone receptors but not integrin αv ß3 blockers. Results suggest that nonpeptide hormones inhibit resveratrol-induced antiproliferation in cancer cells downstream of the interaction between ligand and receptor and ERK1/2 activation to interfere with nuclear COX-2 accumulation. Thus, the surface receptor sites for resveratrol and nonpeptide hormones are distinct and can induce discrete ERK1/2-dependent downstream antiproliferation biological activities. It also indicates the complex pathways by which antiproliferation is induced by resveratrol in various physiological hormonal environments. .


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Estrogens/pharmacology , Signal Transduction/drug effects , Stilbenes/pharmacology , Thyroid Hormones/pharmacology , Animals , Phosphorylation/drug effects , Resveratrol
13.
J Biomed Sci ; 24(1): 51, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28750624

ABSTRACT

BACKGROUND: Obesity and its comorbidities constitute a serious health burden worldwide. Leptin plays an important role in diet control; however, it has a stimulatory potential on cancer cell proliferation. The OB3 peptide, a synthetic peptide, was shown to be more active than leptin in regulating metabolism but with no mitogenic effects in cancer cells. METHODS: In this study, we investigated the proliferative effects, gene expressions and signaling pathways modulated by leptin and OB3 in human ovarian cancer cells. In addition, an animal study was performed. RESULTS: Leptin, but not OB3, induced the proliferation of ovarian cancer cells. Interestingly, OB3 blocked the leptin-induced proliferative effect when it was co-applied with leptin. Both leptin and OB3 activated the phosphatidylinositol-3-kinase (PI3K) signal transduction pathway. In addition, leptin stimulated the phosphorylation of signal transducer and activator of transcription-3 (STAT3) Tyr-705 as well as estrogen receptor (ER)α, and the expression of ERα-responsive genes. Interestingly, all leptin-induced signal activation and gene expressions were blocked by the co-incubation with OB3 and the inhibition of extracellular signal-regulated kinase (ERK)1/2. Coincidently, leptin, but not OB3, increased circulating levels of follicle-stimulating hormone (FSH) which is known to play important roles in the initiation and proliferation of ovarian cancer cells. CONCLUSIONS: In summary, our findings suggest that the OB3 peptide may prevent leptin-induced ovarian cancer initiation and progression by disrupting leptin-induced proliferative signals via STAT3 phosphorylation and ERα activation. Therefore, the OB3 peptide is a potential anticancer agent that might be employed to prevent leptin-induced cancers in obese people.


Subject(s)
Gene Expression Regulation, Neoplastic , Leptin/genetics , Leptin/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/physiopathology , Peptide Fragments/metabolism , Signal Transduction/genetics , Animals , Cell Proliferation/genetics , Female , Humans , Male , Mice, Inbred BALB C , Mice, Nude
14.
Tumour Biol ; 39(6): 1010428317702650, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28639888

ABSTRACT

Actin-binding proteins are proteins that could bind to actin or actin fibers. As a member of actin-binding proteins, Transgelin-2 is expressed in smooth muscle cells and non-smooth muscle cells, and its gene, TAGLN2, is differently expressed in all cells and tissues. The deregulation of Transgelin-2 is considered to be correlated with progression of many kinds of diseases, especially the development of malignant tumors, such as invasion, metastasis, and resistance, yet the function and mechanism of action of Transgelin-2 remain elusive. Therefore, we reviewed the basic characteristics and function of Transgelin-2 and its biological role in various types of diseases in order to provide the theoretical basis for further research and new perspectives on cancer development.


Subject(s)
Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Microfilament Proteins/genetics , Muscle Proteins/genetics , Neoplasms/genetics , Actins/genetics , Humans , Neoplasms/pathology
16.
Eur J Clin Pharmacol ; 73(7): 843-854, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28378058

ABSTRACT

PURPOSE: The substantial variability in the antiplatelet efficacy of clopidogrel has raised major concerns. Molecular epidemiological research suggests that ABCB1 C3435T polymorphism may be associated with clopidogrel response, but results remain controversial. To derive a more precise evaluation of the associations between ABCB1 C3435T polymorphism and the clinical efficacy of clopidogrel, we have conducted a PRISMA-compliant meta-analysis. METHODS: The PubMed and EMBASE databases were searched for eligible studies up to 25 October 2016. The odds ratio (OR), the standard mean difference (SMD) and 95% confidence interval (CI) were applied to assess the strength of the relationship. RESULTS: Overall, 28 related studies involving 23,243 patients were analyzed. No association was found between the ABCB1 polymorphisms and the primary outcome. In the subgroup analysis, the C3435T mutation significantly reduced platelet activity as tested by the LTA assay in the dominant (SMD -0.140, 95% CI -0.272 to -0.009, P = 0.036) and heterozygous (SMD -0.154, 95% CI -0.290 to -0.017, P = 0.027) models, but the result lacked robustness in the sensitivity analysis. A significant association between the C3435T polymorphism and bleeding risk was also observed with low heterogeneity in the dominant (OR 1.805, 95% CI1.124-2.900, P =0.015, I 2 = 0%), homozygous (OR 1.952, 95% CI 1.055-3.611, P = 0.033, I 2 = 13.2%) and heterozygous (OR 1.793, 95% CI 1.091-2.946, P = 0.021, I 2 = 0%) models in Asian patients. CONCLUSIONS: The results of the meta-analysis suggest that ABCB1 C3435T polymorphism may increase the risk of bleeding in Asian patients treated with clopidogrel. The implied relationship needs to be verified in future basic genetic pharmacology studies.


Subject(s)
Coronary Artery Disease/drug therapy , Coronary Artery Disease/genetics , Platelet Aggregation Inhibitors/adverse effects , Ticlopidine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B/genetics , Asian People/genetics , Clopidogrel , Hemorrhage/chemically induced , Hemorrhage/genetics , Humans , Platelet Aggregation Inhibitors/therapeutic use , Polymorphism, Genetic , Risk Factors , Ticlopidine/adverse effects , Ticlopidine/therapeutic use
17.
Int J Oncol ; 50(3): 893-902, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28197640

ABSTRACT

Breast cancer is the leading cause of cancer death among women. Paclitaxel, a mitotic inhibitor, is highly effective in the treatment of breast cancer. However, development of resistance to paclitaxel limits its clinical use. Identifying new compounds and new strategies that are effective against breast cancer, in particular drug-resistant cancer, is of great importance. the aim of the present study was to explore the potential of a next-generation taxoid, SB-T-121205, in modulating the proliferation, migration and invasion of paclitaxel-resistant human breast cancer cells (MCF-7/PTX) and further evaluate the underlying molecular mechanisms. The results of MTT assay showed that SB-T-121205 has much higher potency to human breast cancer cells (MCF-7/S, MCF-7/PTX and MDA-MB-453 cells) than paclitaxel, while that the non-tumorigenic human bronchial epithelial cells (BEAS-2B) were slightly less sensitive to SB-T-121205 than paclitaxel. Flow cytometry and western blot methods revealed that SB-T-121205 induced cell cycle arrest at the G2/M phase and apoptosis in MCF-7/PTX cells through accelerating mitochondrial apoptotic pathway, resulting in reduction of Bcl-2/Bax ratio, as well as elevation of caspase-3, caspase-9, and poly(ADP-ribose) polymerase (PARP) levels. Moreover, SB-T-121205 changed epithelial-mesenchymal transition (EMT) property, and suppressed migration and invasion abilities of MCF-7/PTX cells. Additionally, SB-T-121205 exerted antitumor activity by inhibiting the transgelin 2 and PI3K/Akt pathway. These findings indicate that SB-T-121205 is a potent antitumor agent that promotes apoptosis and also recedes migration/invasion abilities of MCF-7/PTX cells by restraining the activity of transgelin 2 and PI3K/Akt, as well as mitochondrial apoptotic pathway. Such results suggest a potential clinical value of SB-T-121205 in breast cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Taxoids/pharmacology , Breast Neoplasms/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , MCF-7 Cells , Microfilament Proteins/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Neoplasm Invasiveness/pathology , Paclitaxel/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Spheroids, Cellular , Tumor Cells, Cultured , bcl-2-Associated X Protein/metabolism
18.
J Drug Target ; 25(3): 255-263, 2017 03.
Article in English | MEDLINE | ID: mdl-27718638

ABSTRACT

Patient SE translation (SET) is a carcinogen in facilitating cellular growth and proliferation, and promoting tumorigenesis and metastasis. The present study was to investigate the resistance mechanisms associated with SET in paclitaxel-induced human breast cancer cells. The different expressions of SET, ATP-binding cassette (ABC) transporters and PI3K/Akt pathway between paclitaxel sensitive MCF-7/S and paclitaxel resistant MCF-7/PTX cells were identified using western blotting. We adopted plasmid transfection to upregulate SET in MCF-7/S cells and a novel SET antagonist COG112 to decrease SET in MCF-7/PTX cells. Subsequently, cell viability to paclitaxel was assessed by MTT assay and cell apoptosis was analyzed by flow cytometry. We found that levels of SET, ABC transporters and PI3K/Akt pathway were elevated in MCF-7/PTX. Upregulation of SET in MCF-7/S cells expressed resistant to paclitaxel and decreased cell apoptosis. Moreover, overexpression of SET promoted the mRNA and protein level of ABC transporters and PI3K/Akt signal pathway in MCF-7/S cells. Conversely, decreased level of SET by COG112 not only significantly sensitized MCF-7/PTX cells to paclitaxel, but also enhanced paclitaxel-induced cell apoptosis. Additionally, the levels of the ABC transporters and PI3K/Akt signal pathway were also reduced in the COG112-treated MCF-7/PTX cells. The above results demonstrated that SET was associated with paclitaxel resistance in MCF-7/PTX cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Histone Chaperones/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , Blotting, Western , DNA-Binding Proteins , Down-Regulation , Drug Resistance, Neoplasm , Enzyme Activation , Female , Flow Cytometry , Humans , MCF-7 Cells , Real-Time Polymerase Chain Reaction
19.
Oncotarget ; 8(15): 24237-24249, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-27458161

ABSTRACT

Ovarian cancer is the leading cause of death in gynecological diseases. Thyroid hormone promotes proliferation of ovarian cancer cells via cell surface receptor integrin αvß3 that activates extracellular regulated kinase (ERK1/2). However, the mechanisms are still not fully understood. Thyroxine (T4) at a physiologic total hormone concentration (10-7 M) significantly increased proliferating cell nuclear antigen (PCNA) abundance in these cell lines, as did 3, 5, 3'-triiodo-L-thyronine (T3) at a supraphysiologic concentration. Thyroid hormone (T4 and T3) treatment of human ovarian cancer cells resulted in enhanced activation of the Ras/MAPK(ERK1/2) signal transduction pathway. An MEK inhibitor (PD98059) blocked hormone-induced cell proliferation but not ER phosphorylation. Knock-down of either integrin αv or ß3 by RNAi blocked thyroid hormone-induced phosphorylation of ERK1/2. We also found that thyroid hormone causes elevated phosphorylation and nuclear enrichment of estrogen receptor α (ERα). Confocal microscopy indicated that both T4 and estradiol (E2) caused nuclear translocation of integrin αv and phosphorylation of ERα. The specific ERα antagonist (ICI 182,780; fulvestrant) blocked T4-induced ERK1/2 activation, ERα phosphorylation, PCNA expression and proliferation. The nuclear co-localization of integrin αv and phosphorylated ERα was inhibited by ICI. ICI time-course studies indicated that mechanisms involved in T4- and E2-induced nuclear co-localization of phosphorylated ERα and integrin αv are dissimilar. Chromatin immunoprecipitation results showed that T4-induced binding of integrin αv monomer to ERα promoter and this was reduced by ICI. In summary, thyroid hormone stimulates proliferation of ovarian cancer cells via crosstalk between integrin αv and ERα, mimicking functions of E2.


Subject(s)
Estrogen Receptor alpha/metabolism , Integrin alphaVbeta3/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ovarian Neoplasms/pathology , Protein Binding
20.
Fundam Clin Pharmacol ; 31(3): 311-318, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28012206

ABSTRACT

Invasive pulmonary aspergillosis (IPA), most caused by Aspergillus fumigatus, is a serious life-threatening infection in immunocompromised patients. Voriconazole is used to prevent and treat IPA. However, little is known about the pharmacological characteristics of voriconazole in pulmonary epithelial cells, which are the target site for the prophylaxis and early treatment of IPA. The aim of the study was to evaluate the kinetics and activity of voriconazole against A. fumigatus in A549 cells. High-performance liquid chromatography/tandem mass spectrometry and time-kill method were used to study the cellular pharmacokinetic and pharmacodynamics of voriconazole. Voriconazole exerted a concentration-dependent toxic effect on A549 cells and could penetrate into cells, reaching plateau concentrations of 1.14 ± 0.64, 3.72 ± 1.38 and 6.36 ± 0.95 ng/mg protein after A549 cells were exposed to voriconazole at extracellular concentrations of 2, 8 and 16 mg/L for 2 h, respectively. The efflux of voriconazole was rapid, with a half-life of 10.2 min. Voriconazole can decrease the A. fumigatus conidia invade cells, and the number of viable A. fumigatus conidia in cells can be decreased 2.1- to 20.6-fold when A549 cells were cultured in medium containing voriconazole. After 24-h incubation, 75.6% and 80.5% of intracellular A. fumigatus were killed when extracellular voriconazole concentration was 8 and 16 mg/L, respectively. This study illustrated a new application for the prophylaxis and early treatment of IPA from the cellular pharmacokinetics and pharmacodynamics and emphasized the importance of monitoring concentrations of voriconazole in epithelial lining fluid in immunocompromised patients receiving voriconazole therapy.


Subject(s)
Aspergillus fumigatus/drug effects , Epithelial Cells/drug effects , Invasive Pulmonary Aspergillosis/drug therapy , Lung/drug effects , Voriconazole/pharmacology , Voriconazole/pharmacokinetics , A549 Cells , Antibiotic Prophylaxis/methods , Antifungal Agents/pharmacokinetics , Antifungal Agents/pharmacology , Cell Line, Tumor , Epithelial Cells/microbiology , Half-Life , Humans , Invasive Pulmonary Aspergillosis/microbiology , Kinetics , Lung/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL