Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Chemosphere ; 193: 1164-1171, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29874745

ABSTRACT

Ammonium (NH4+) would influence the uptake and translocation of Cd and Zn to mediate their interactions in wheat. Thus, the effects of NH4+ on Cd and Zn uptake, translocation, subcellular distributions and Cd chemical forms in dwarf polish wheat (DPW, Triticum polonicum L.) under Cd, Zn and Cd + Zn stresses with lack or supply of NH4+ was investigated. The biomasses of root and shoot were reduced by NH4+. NH4+ enhanced Cd and Zn uptakes, but inhibited their translocations. Under lack and supply of NH4+, Zn inhibited Cd uptakes, but promoted Cd translocations. Meanwhile, NH4+ reinforced the inhibition of Cd uptake and the promotion of Cd translocation caused by Zn. Cd inhibited Zn uptake and promoted Zn translocation under lack of NH4+. Meanwhile, Cd slightly reduced the Zn uptake, but did not affect the translocation under supply of NH4+. Therefore, NH4+ alleviated the inhibition of Zn uptake and partly reduced the promotion of Zn translocation stimulated by Cd. NH4+ and Zn changed the subcellular distributions and chemical forms of Cd. NH4+ and Cd also influenced the subcellular distributions of Zn. The changed subcellular distributions and chemical forms were associated with Cd and Zn uptakes and translocations, which physiologically revealed and illustrated NH4+ participates in Cd/Zn interactions.


Subject(s)
Ammonium Compounds/chemistry , Cadmium/chemistry , Seedlings/drug effects , Triticum/drug effects , Zinc/chemistry , Plant Roots/drug effects
2.
Cancer Lett ; 422: 56-69, 2018 05 28.
Article in English | MEDLINE | ID: mdl-29458143

ABSTRACT

Suppressor of variegation 3-9 homolog 2 (SUV39H2) is a member of the SUV39H subfamily of lysine methyltransferases. Its role in colorectal cancer (CRC) proliferation and metastasis has remained unexplored. Here, we determined that SUV39H2 was upregulated in CRC tissues compared with that in adjacent non-neoplastic tissues. Further statistical analysis revealed that high SUV39H2 expression was strongly associated with distant metastasis (P = 0.016) and TNM stage (P = 0.038) and predicted a shorter overall survival (OS; P = 0.018) and progression-free survival (PFS; P = 0.018) time for CRC patients. Both in vitro and in vivo assays demonstrated that ectopically expressed SUV39H2 enhanced CRC proliferation and metastasis, while SUV39H2 knockdown inhibited CRC proliferation and metastasis. A molecular screen of SUV39H2 targets found that SUV39H2 negatively regulated the expression of SLIT guidance ligand 1 (SLIT1). Moreover, rescue assays suggested that SLIT1 could antagonize the function of SUV39H2 in CRC. Mechanistic studies indicated that SUV39H2 can directly bind to the SLIT1 promoter, suppressing SLIT1 transcription by catalyzing histone H3 lysine 9 (H3K9) tri-methylation. In summary, we propose that SUV39H2 can predict CRC patient prognosis and stimulate CRC malignant phenotypes via SLIT1 promoter tri-methylation.


Subject(s)
Colorectal Neoplasms/pathology , DNA Methylation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Nerve Tissue Proteins/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Histones/metabolism , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Neoplasm Transplantation , Promoter Regions, Genetic , Survival Analysis , Up-Regulation
3.
Int J Cancer ; 142(7): 1379-1391, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29164615

ABSTRACT

Rho guanine nucleotide exchange factors (RhoGEFs) are proteins that activate Rho GTPases in response to extracellular stimuli and regulate various biologic processes. ARHGEF19, one of RhoGEFs, was reported to activate RhoA in the Wnt-PCP pathway controlling convergent extension in Xenopus gastrulation. The goal of our study was to identify the role and molecular mechanisms of ARHGEF19 in the tumorigenesis of non-small cell lung cancer (NSCLC). ARHGEF19 expression was significantly elevated in NSCLC tissues, and ARHGEF19 levels were significantly associated with lymph node status, distant metastasis and TNM stage; Patients with high ARHGEF19 levels had poor overall survival (OS) and progression-free survival (PFS). Our investigations revealed that ARHGEF19 overexpression promoted the cell proliferation, invasion and metastasis of lung cancer cells, whereas knockdown of this gene inhibited these processes. Mechanistically, ARHGEF19 activated the mitogen-activated protein kinase (MAPK) pathway in a RhoA-independent manner: ARHGEF19 interacted with BRAF and facilitated the phosphorylation of its downstream kinase MEK1/2; both the Dbl homology (DH) and Pleckstrin homology (PH) domains of ARHGEF19 were indispensable for the phosphorylation of MEK1/2. Furthermore, downregulation of miR-29b was likely responsible for the increased expression of ARHGEF19 in lung cancer tissues and, consequently, the abnormal activation of MAPK signaling. These findings suggest that ARHGEF19 upregulation, due to the low expression of miR-29 in NSCLC tissues, may play a crucial role in NSCLC tumorigenesis by activating MAPK signaling. ARHGEF19 could serve as a negative prognostic marker as well as a therapeutic target for NSCLC patients.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Gene Expression Regulation, Neoplastic/physiology , Guanine Nucleotide Exchange Factors/metabolism , Lung Neoplasms/pathology , Animals , Area Under Curve , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Disease-Free Survival , Female , Guanine Nucleotide Exchange Factors/genetics , Heterografts , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , MicroRNAs/biosynthesis , MicroRNAs/genetics , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , ROC Curve , Sensitivity and Specificity , Signal Transduction/physiology
4.
Cell Death Dis ; 8(6): e2874, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28617432

ABSTRACT

Concurrent/adjuvant cisplatin-based chemoradiotherapy is regarded as the standard of treatment for locoregionally advanced nasopharyngeal carcinoma (NPC). However, patients who do not respond to cisplatin suffer, rather than benefit, from chemotherapy treatment. The goal of this study was to identify molecules involved in cisplatin resistance and to clarify their molecular mechanisms, which would help in the discovery of potential therapeutic targets and in developing a personalized and precise treatment approach for NPC patients. We previously generated a cisplatin-sensitive NPC cell line, S16, from CNE2 cells and found that eIF3a, ASNS and MMP19 are upregulated in S16 cells, which contributes to their cisplatin sensitivity. In this study, we found that BST2 is downregulated in cisplatin-sensitive S16 cells compared with CNE2 cells. Knockdown of BST2 in NPC cells sensitized their response to cisplatin and promoted cisplatin-induced apoptosis, whereas exogenous overexpression of BST2 increased their cisplatin resistance and inhibited cisplatin-induced apoptosis. Further investigation demonstrated that BST2-mediated cisplatin resistance depended on the activation of the NF-κB signaling pathway and consequent upregulation of anti-apoptotic genes, such as Bcl-XL and livin. Moreover, an analysis of clinical data revealed that a high BST2 level might serve as an independent indicator of poor prognosis in patients with locally advanced NPC treated with platinum-based chemoradiotherapy. These findings suggest that BST2 likely mediates platinum resistance in NPC, offering guidance for personalized and precise treatment strategies for patients with NPC.


Subject(s)
Antigens, CD/metabolism , Carcinoma/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm , NF-kappa B/metabolism , Nasopharyngeal Neoplasms/drug therapy , Adult , Aged , Animals , Apoptosis , Carcinoma/metabolism , Cell Line, Tumor , Down-Regulation , Female , GPI-Linked Proteins/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Neoplasm Transplantation , Prognosis , Signal Transduction
5.
J Pathol ; 240(3): 352-365, 2016 11.
Article in English | MEDLINE | ID: mdl-27555280

ABSTRACT

Uncontrolled growth and distant metastasis are hallmarks of colorectal cancer (CRC), but the mechanisms are poorly understood. Olfactomedin 1 (OLFM1), a member of the olfactomedin domain-containing protein family, plays an important role in the development of neurogenic tissues. Recently, OLFM1 deregulation was frequently observed in several cancers, and it was induced in colon cell lines after treatment with the demethylating agent 5-aza-2'-deoxycytidine. However, the function of OLFM1 in CRC remains unknown. In this study, we reanalysed published microarray data and found that OLFM1 was significantly down-regulated in primary CRC samples compared to adjacent non-cancerous tissues. The results of immunohistochemistry indicated that decreased OLFM1 expression was significantly associated with lymph node status (p = 0.023), distant metastasis (p < 0.001), and AJCC/TNM stage (p = 0.013), and CRC patients with low OLFM1 expression had consistently poor overall survival (OS; p < 0.001) and progression-free survival (PFS; p < 0.001). Further analysis demonstrated that OLFM1 was epigenetically silenced in CRC tissues and cell lines via promoter hypermethylation. Overexpression and knockdown of OLFM1 attenuated and increased, respectively, CRC cells' proliferation, migration, and invasion in vitro and metastasis to the lung and liver in vivo. Mechanistically, the promotion of growth and metastasis of CRC cells by silencing of OLFM1 was associated with the activation of the non-canonical NF-κB signalling pathway. OLFM1 interacted with NF-κB-inducing kinase (NIK; MAP3K14) and repressed the phosphorylation of its downstream substrate Ikappa B kinase alpha (IKKα). OLFM1 expression was negatively correlated with the phosphorylation level of IKKα in CRC tissue samples. Knockdown of NIK impaired the ability of OLFM1 to repress NF-κB signalling, cell growth or migration. Thus, OLFM1 may be a valuable biomarker and therapeutic target for CRC patients. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Colorectal Neoplasms/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Liver Neoplasms/secondary , Lung Neoplasms/secondary , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Biomarkers/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , DNA Methylation , Decitabine , Disease-Free Survival , Down-Regulation , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Humans , Liver Neoplasms/pathology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NF-kappa B/genetics , Prognosis , Protein Serine-Threonine Kinases/genetics , Signal Transduction , NF-kappaB-Inducing Kinase
6.
Oncotarget ; 6(29): 28478-90, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26156020

ABSTRACT

In this study, we tried to explore if xeroderma pigmentosum complementation group-A (XPA) expression is likely a prognostic prediction factor for locally advanced nasopharyngeal carcinoma (NPC) patients treated with platinum-based chemoradiotherapy, which was considered to bring chemotherapy-related severe toxicity compared with radiotherapy alone. Firstly, MTT assay revealed that downregulating XPA expression in NPC HONE1 and CNE1 cells decreased IC50 of cisplatin and sensitized cells to cisplatin. XPA expression was detected by immunohistochemistry in cancer tissues from locally advanced NPC patients treated with platinum-based chemoradiotherapy. The relationships between XPA expression and clinicopathologic features, overall survival and progression-free survival of patients were evaluated. The results showed that XPA expression was not associated with clinicopathologic parameters, but was likely an independent prognostic factor for patient survival. High XPA level predicts a poor prognosis, and the prediction values were higher in subgroups of younger, higher EBV antibody titer, or treated with concurrent chemoradiotherapy. Combining XPA levels and T/N classifications, we successfully classified these patients into low, medium and high risk groups for platinum-based chemoradiotherapy. These findings suggest that XPA levels may be a potential predictor of prognosis in locally advanced NPC patients treated with platinum-based chemoradiotherapy, and helpful for selecting patients likely to need and benefit from this treatment in future.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/therapy , Xeroderma Pigmentosum Group A Protein/metabolism , Blotting, Western , Carcinoma , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy/methods , Cisplatin/administration & dosage , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Immunohistochemistry , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Prognosis , Proportional Hazards Models , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Xeroderma Pigmentosum Group A Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...