BackgroundTreatment options for metastatic castrate-resistant prostate cancer (mCRPC) are limited and typically are centered on docetaxel-based chemotherapy. We previously reported that elevated miR-375 levels were significantly associated with poor overall survival of mCRPC patients. In this study, we evaluated if miR-375 induced chemo-resistance to docetaxel through regulating target genes associated with drug resistance.MethodsWe first compared miR-375 expression level between prostate cancer tissues and normal prostate tissues using data from The Cancer Genome Atlas (TCGA). To examine the role of miR-375 in docetaxel resistance, we transfected miR-375 using a pre-miRNA lentiviral vector and examined the effects of exogenously overexpressed miR-375 on cell growth in two prostate cancer cell lines, DU145 and PC-3. To determine the effect of overexpressed miR-375 on tumor growth and chemo-resistance in vivo, we injected prostate cancer cells overexpressing miR-375 into nude mice subcutaneously and evaluated tumor growth rate during docetaxel treatment. Lastly, we utilized qRT-PCR and Western blot assay to examine two miR-375 target genes, SEC23A and YAP1, for their expression changes after miR-375 transfection.ResultsBy examining 495 tumor tissues and 52 normal tissues from TCGA data, we found that compared to normal prostate, miR-375 was significantly overexpressed in prostate cancer tissues (8.45-fold increase, p value = 1.98E-23). Docetaxel treatment induced higher expression of miR-375 with 5.83- and 3.02-fold increases in DU145 and PC-3 cells, respectively. Interestingly, miR-375 appeared to play a dual role in prostate cancer proliferation. While miR-375 overexpression caused cell growth inhibition and cell apoptosis, elevated miR-375 also significantly reduced cell sensitivity to docetaxel treatment in vitro, as evidenced by decreased apoptotic cells. In vivo xenograft mouse study showed that tumors with increased miR-375 expression were more tolerant to docetaxel treatment, demonstrated by greater tumor weight and less apoptotic cells in miR-375 transfected group when compared to empty vector control group. In addition, we examined expression levels of the two miR-375 target genes (SEC23A and YAP1) and observed significant reduction in the expression at both protein and mRNA levels in miR-375 transfected prostate cancer cell lines. TCGA dataset analysis further confirmed the negative correlations between miR-375 and the two target genes (r = −0.62 and −0.56 for SEC23A and YAP1, respectively; p < 0.0001).ConclusionsmiR-375 is involved in development of chemo-resistance to docetaxel through regulating SEC23A and YAP1 expression. Our results suggest that miR-375 or its target genes, SEC23A or YAP1, might serve as potential predictive biomarkers to docetaxel-based chemotherapy and/or therapeutic targets to overcome chemo-resistance in mCRPC stage.Electronic supplementary materialThe online version of this article (doi:10.1186/s12943-016-0556-9) contains supplementary material, which is available to authorize...
Five platinum(II) 1,4,7-trithiacyclononane (ttcn) complexes with bidentate-substituted 2,2'-bipyridine ligands have been prepared and structurally characterized: [Pt(bpy)(ttcn)](PF6)2 (bpy = 2,2'-bipyridine), triclinic, P1, a = 10.2529(3) A, b = 10.7791(3) A, c = 10.7867(3) A, alpha = 83.886(1) degrees, beta = 87.565(1) degrees, gamma = 84.901(1), V = 1179.99(6) A3, Z = 2; [Pt(4,4'-dmbpy)(ttcn)](PF6)2 x CH3CN x H2O (4,4'-dmbpy = 4,4'-dimethyl-2,2'-bipyridine), triclinic, P1, a = 10.1895(3) A, b = 11.8566(4) A, c = 13.1004(4) A, alpha = 77.345(1) degrees, beta = 79.967(1) degrees, gamma = 72.341(1) degrees, V = 1461.56(8) A3, Z = 2; [Pt(5,5'-dmbpy)(ttcn)](PF6)2 (5,5'-dmbpy = 5,5'-dimethyl-2,2'-bipyridine), triclinic, P1, a = 10.6397(4) A, b = 10.8449(4) A, c = 11.2621(4) A, alpha = 90.035(1) degrees, beta = 98.061(1) degrees, gamma = 91.283(1) degrees, V = 1286.32(8) A3, Z = 2; [Pt(dbbpy)(ttcn)](PF6)2 x CH3NO2 (dbbpy = 4,4'-di-tert-butyl-2,2'-bipyridine), triclinic, P1, a = 11.5422(7) A, b = 11.6100(7) A, c = 13.6052(9) A, alpha = 85.902(1) degrees, beta = 89.675(1) degrees, gamma = 74.942(1) degrees, V = 1755.90(19) A3, Z = 2; and [Pt(dtfmbpy)(ttcn)](PF6)2 x CH3CN (dtfmbpy = 5,5'-di-trifluoromethyl-2,2'-bipyridine): monoclinic, P2(1)/c, a = 13.1187(9) A, b = 20.9031(15) A, c = 11.3815(8) A, beta = 105.789(2) degrees, V = 3003.3(4) A3, Z = 4. For each salt, the platinum(II) center of the cation is bonded to two nitrogen atoms of the chelating diimine and two sulfur atoms of the thioether macrocycle. The third sulfur atom of ttcn forms a long apical interaction with the metal center (2.84-2.97 A), resulting in a flattened square pyramid structure. An examination of these and 17 other structures of platinum(II) ttcn complexes reveals a correlation between the apical Pt...S distance and the donor properties of the ancillary ligands, suggesting a means for using variations in ligand electronic properties to tune molecular structure. The room-temperature absorption spectra in acetonitrile solution show a broad and comparatively low-energy MLCT band maximizing near approximately 390 nm for the bpy and dialkyl-substituted bipyridyl derivatives. The maximum is dramatically red-shifted to 460 nm in the spectrum of the dtfmbpy complex as a result of the electron-withdrawing properties of the -CF(3) groups. The 3:1 EtOH/MeOH 77 K glassy solution emission spectra exhibit low-energy emission bands (lambdamax, 570-645 nm), tentatively assigned as originating from a lowest, predominantly spin-forbidden MLCT excited state that is stabilized by apical Pt...S interactions.
Androgen Receptor (AR) -signaling plays a critical role in the development and progression of prostate cancer. Tumor microvasculature contributes to continual exposure of prostate cancer cells to hypoxia-reoxygenation, however, the role of hypoxia-reoxygenation in prostate cancer progression and modulation of AR signaling is not understood. In the present study, we evaluated the effects of hypoxia-reoxygenation in LNCaP cells, a line of hormone responsive human prostate cancer cells. Our results demonstrate that hypoxia-reoxygenation resulted in increased survival, higher clonogenicity, and enhanced invasiveness of these cells. Moreover, hypoxia-reoxygenation was associated with an increased AR activity independent of androgens as well as increased Hypoxia Inducible Factor (HIF-1α) levels and activity. We also observed that the activation of p38 MAP kinase pathway was an early response to hypoxia, and inhibition of p38 MAP kinase pathway by variety of approaches abolished hypoxia-reoxygenation induced increased AR activity as well as increased survival, clonogenicity, and invasiveness. These results demonstrate a critical role for hypoxia induced p38 MAP kinase pathway in androgen independent AR activation in prostate cancer cells, and suggest that hypoxia re-oxygenation may select for aggressive androgen-independent prostate cancer phenotype.
This analytical method can be used for quantitating contraceptive steroid levels in women for monitoring systemic exposure to determine drug interactions, nonadherence, misreporting and proper dosing.
Recent genome-wide association studies have identified variations in the recombination repair gene, RAD52, that are associated with increased lung cancer risk, and particularly with the development of lung squamous cell carcinomas (LUSC). LUSC development is strongly associated with smoking. DNA repair is increased in the lung tissues of smokers, presumably because of ongoing DNA damage from exposure to tobacco smoke. A key player in the DNA damage response, RAD52 plays a role in DNA strand exchange and annealing during homologous recombination (HR) in mammalian cells. In this study, we discovered two cis-expression quantitative trait loci (eQTL) SNPs in the RAD52 gene that are associated with its expression and are also associated with LUSC risk. In addition, we report that amplification of the genomic region 12p13.33, which contains the RAD52 gene, is significantly associated with the development of LUSC in the TCGA database and that somatic overexpression of RAD52 was confirmed to be significant in LUSC tumors from our own patient cohort. Consistent with these genetic findings, we demonstrate that blockade of Rad52 slows cell growth and induces senescence in mouse bronchial epithelial cells. In contrast, overexpression of Rad52 leads to an increased rate of cell proliferation. We show that depletion of Rad52 in mouse lung tumor cells alters cell cycle distribution and increases DNA damage accumulation associated with increased tumor cell death. Our genetic and functional data implicate RAD52 as a significant determinant of risk in the development of LUSC.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.