Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Drug Dev Res ; 84(7): 1537-1552, 2023 11.
Article in English | MEDLINE | ID: mdl-37571819

ABSTRACT

FAM64A is a mitogen-induced regulator of the metaphase and anaphase transition. Here, we found that FAM64A messenger RNA (mRNA) and protein expression levels were higher in gastric cancer tissue than in normal mucosa (p < .05). FAM64A methylation was negatively correlated with FAM64A mRNA expression (p < .05). The differentially expressed genes of FAM64A were mainly involved in digestion, potassium transporting or exchanging ATPase, contractile fibers, endopeptidase, and pancreatic secretion (p < .05). The FAM64A-related genes were principally categorized into ubiquitin-mediated proteolysis, cell cycle, chromosome segregation and mitosis, microtubule binding and organization, metabolism of amino acids, cytokine receptors, lipid droplet, central nervous system, and collagen trimer (p < .05). FAM64A protein expression was lower in normal gastric mucosa than intestinal metaplasia, adenoma, and primary cancer (p < .05), negatively correlated with older age, T stage, lymphatic and venous invasion, tumor, node, metastasis stage, and dedifferentiation (p < .05), and associated with a favorable overall survival of gastric cancer patients. FAM64A overexpression promoted proliferation, antiapoptosis, migration, invasion, and epithelial-mesenchymal transition via the EGFR/Akt/mTOR/NF-κB, while the opposite effect was observed for FAM64A knockdown. FAM64A also induced chemoresistance directly or indirectly through lipid droplet formation via ING5. These results suggested that upregulation of FAM64A expression might induce aggressive phenotypes, leading to gastric carcinogenesis and its subsequent progression. Thus, FAM64A could be regarded as a prognosis biomarker and a target for gene therapy.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Lipid Droplets/metabolism , Lipid Droplets/pathology , Gene Expression Regulation, Neoplastic , Biomarkers , Cell Proliferation/genetics , RNA, Messenger , Genetic Therapy , Cell Line, Tumor , Cell Movement , Prognosis
2.
Front Cell Dev Biol ; 10: 1012179, 2022.
Article in English | MEDLINE | ID: mdl-36425530

ABSTRACT

As a Class II tumor suppressor, ING5 contains nuclear localization signal, plant homeodomain, novel conserved region, and leucine zipper-like domains. ING5 proteins form homodimer into a coil-coil structure, and heterodimers with ING4, histone H3K4me3, histone acetyltransferase (HAT) complex, Tip60, Cyclin A1/CDK2, INCA1 and EBNA3C for the transcription of target genes. The acetylated proteins up-regulated by ING5 are preferentially located in nucleus and act as transcription cofactors, chromatin and DNA binding functions, while those down-regulated by ING5 mostly in cytoplasm and contribute to metabolism. ING5 promotes the autoacetylation of HAT p300, p53, histone H3 and H4 for the transcription of downstream genes (Bax, GADD45, p21, p27 and so forth). Transcriptionally, YY1 and SRF up-regulate ING5 mRNA expression by the interaction of YY1-SRF-p53-ING5 complex with ING5 promoter. Translationally, ING5 is targeted by miR-196, miR-196a, miR-196b-5p, miR-193a-3p, miR-27-3p, miR-200b/200a/429, miR-1307, miR-193, miR-222, miR-331-3p, miR-181b, miR-543 and miR-196-b. ING5 suppresses proliferation, migration, invasion and tumor growth of various cancer cells via the suppression of EGFR/PI3K/Akt, IL-6/STAT3, Akt/NF-κB/NF-κB/MMP-9 or IL-6/CXCL12 pathway. ING5-mediated chemoresistance is closely linked to anti-apoptosis, overexpression of chemoresistant genes, the activation of PI3K/Akt/NF-κB and Wnt/ß-catenin signal pathways. Histologically, ING5 abrogation in gastric stem-like and pdx1-positive cells causes gastric dysplasia and cancer, and conditional ING5 knockout in pdx1-positive and gastric chief cells increases MNU-induced gastric carcinogenesis. Intestinal ING5 deletion increases AOM/DSS- induced colorectal carcinogenesis and decreases high-fat-diet weight. The overexpression and nucleocytoplasmic translocation of ING5 are seen during carcinogenesis, and ING5 expression was inversely associated with aggressive behaviors and poor prognosis in a variety of cancers. These findings indicated that ING5 might be used for a molecular marker for carcinogenesis and following progression, and as a target for gene therapy if its chemoresistant function might be ameliorated.

3.
Front Oncol ; 12: 744886, 2022.
Article in English | MEDLINE | ID: mdl-35350574

ABSTRACT

JC polyoma virus (JCPyV) is a ubiquitous polyoma virus that infects the individual to cause progressive multifocal leukoencephalopathy and malignancies. Here, we found that T-antigen knockdown suppressed proliferation, glycolysis, mitochondrial respiration, migration, and invasion, and induced apoptosis and G2 arrest. The reverse was true for T-antigen overexpression, with overexpression of Akt, survivin, retinoblastoma protein, ß-catenin, ß-transducin repeat-containing protein (TRCP), and inhibitor of growth (ING)1, and the underexpression of mammalian target of rapamycin (mTOR), phosphorylated (p)-mTOR, p-p38, Cyclin D1, p21, vascular endothelial growth factor (VEGF), ING2, and ING4 in hepatocellular and pancreatic cancer cells and tissues. In lens tumor cells, T antigen transcriptionally targeted viral carcinogenesis, microRNAs in cancer, focal adhesion, p53, VEGF, phosphoinositide 3 kinase-Akt, and Forkhead box O signaling pathways, fructose and mannose metabolism, ribosome biosynthesis, and choline and pyrimidine metabolism. At a metabolomics level, it targeted protein digestion and absorption, aminoacryl-tRNA biosynthesis, biosynthesis of amino acids, and the AMPK signal pathway. At a proteomic level, it targeted ribosome biogenesis in eukaryotes, citrate cycle, carbon metabolism, protein digestion and absorption, aminoacryl-tRNA biosynthesis, extracellular-matrix-receptor interaction, and biosynthesis of amino acids. In lens tumor cells, T antigen might interact with various keratins, ribosomal proteins, apolipoproteins, G proteins, ubiquitin-related proteins, RPL19, ß-catenin, ß-TRCP, p53, and CCAAT-enhancer-binding proteins in lens tumor cells. T antigen induced a more aggressive phenotype in mouse and human cancer cells due to oncogene activation, inactivation of tumor suppressors, and disruption of metabolism, cell adhesion, and long noncoding RNA-microRNA-target axes.

4.
Mol Med Rep ; 25(1)2022 Jan.
Article in English | MEDLINE | ID: mdl-34779504

ABSTRACT

Recently accumulated evidence has indicated that the nucleomembrane shuttling of cellular proteins is common, which provides new insight into the subcellular translocation and biological functions of proteins synthesized in the cytoplasm. The present study aimed to clarify the trafficking of proteins between the plasma membrane and nucleus. These proteins primarily consist of transmembrane receptors, membrane adaptor proteins, adhesive proteins, signal proteins and nuclear proteins, which contribute to proliferation, apoptosis, chemoresistance, adhesion, migration and gene expression. The proteins frequently undergo cross­talk, such as the interaction of transmembrane proteins with signal proteins. The transmembrane proteins undergo endocytosis, infusion into organelles or proteolysis into soluble forms for import into the nucleus, while nuclear proteins interact with membrane proteins or act as receptors. The nucleocytosolic translocation involves export or import through nuclear membrane pores by importin or exportin. Nuclear proteins generally interact with other transcription factors, and then binding to the promoter for gene expression, while membrane proteins are responsible for signal initiation by binding to other membrane and/or adaptor proteins. Protein translocation occurs in a cell­specific manner and is closely linked to cellular biological events. The present review aimed to improve understanding of cytosolic protein shuttling between the plasma membrane and nucleus and the associated signaling pathways.


Subject(s)
Cell Membrane/metabolism , Nuclear Envelope/metabolism , Protein Transport/physiology , Active Transport, Cell Nucleus/physiology , Adaptor Proteins, Signal Transducing/metabolism , Biological Transport/physiology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , Humans , Nuclear Proteins/metabolism , Signal Transduction/physiology
5.
Mol Cell Biochem ; 476(4): 1741-1749, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33428060

ABSTRACT

Acute kidney injury (AKI) occurs in 30%-70% of critically ill patients. Multiple organ failure (MOF), which is most often secondary to hypotension and septicemia, is a global public health problem. The prognosis of patients is poor. Currently, there is no specific therapeutic method. Finding new therapeutic targets is significant to improve the prognosis of AKI patients. This study explores expressions and related mechanisms of miR-212 and Kruppel-like factor 4 (KLF4) in rats with AKI. Sixty Wistar rats were randomly divided into 6 groups: Control group, sham operation group, model group, miR-212-agomir group, miR-212-antagomir group, miR-212-agomir+APTO-253 (joint group), n = 10. The expressions of miR-212, KLF4, inflammatory factors [tumor necrosis factor α (TNF-α), interleukin 6 (IL-6)], oxidative stress factors [superoxide dismutase (SOD), malondialdehyde (MDA)], and apoptosis-related proteins (bax, bcl-2) in renal tissue of rats were detected, and the relationship between miR-212 and KLF4 and the severity of AKI in rats were analyzed. The expression level of miR-212 increased (P < 0.05) and the expression level of KLF4 decreased (P < 0.05) in renal tissue of rats with AKI. miR-212 was negatively correlated with KLF4 expression (P < 0.05). MiR-212 was positively correlated with expressions of TNF-α, IL-6, MDA, and bax (P < 0.05), negatively correlated with expressions of SOD and bcl-2 (P < 0.05), KLF4 was negatively correlated with expressions of TNF-α, IL-6, MDA and bax (P < 0.05), and positively correlated with expressions of SOD and bcl-2 (P < 0.05). MiR-212 mimics can inhibit the luciferase activity of Wt-KLF4 (P < 0.05), and miR-212 inhibitor can promote the luciferase activity of Wt-KLF4 (P < 0.05). Down-regulation of miR-212 plays a protective role by targeting up-regulation of KLF4 to inhibit renal tissue inflammation, oxidative stress, and apoptosis in rats with AKI, which may be a potential target for clinical treatment of AKI in the future.


Subject(s)
Acute Kidney Injury/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/biosynthesis , MicroRNAs/biosynthesis , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Animals , Disease Models, Animal , Kruppel-Like Factor 4 , Rats , Rats, Wistar
6.
Front Oncol ; 10: 598192, 2020.
Article in English | MEDLINE | ID: mdl-33330092

ABSTRACT

BTG (B-cell translocation gene) could inhibit cell proliferation, metastasis, and angiogenesis and regulate cell cycle progression and differentiation in a variety of cancer cell types. To clarify the role of BTG1 in invasion and metastasis, its expression was compared with the clinicopathological parameters of colorectal cancer by bioinformatics and immunohistochemical analyses. We also overexpressed BTG1 in HCT-15 cells and examined its effects on adhesion, migration, and metastasis with their related molecules screened. BTG1 mRNA expression was negatively correlated with its promoter methylation in colorectal cancer (P < 0.05). Among them, cg08832851 and cg05819371 hypermethylation and mRNA expression of BTG1 were positively related with poor prognosis of the colorectal cancer patients (P < 0.05). BTG1 expression was found to positively correlate with depth of invasion, venous invasion, lymph node metastasis, distant metastasis, and TNM staging of colorectal cancer (P < 0.05) but negatively with serum levels of CEA and CA19-9 (P < 0.05). According to the TCGA database, BTG1 mRNA expression was lower in well-, moderately, and poorly differentiated than mucinous adenocarcinomas and positively correlated with ras or BRAF mutation (P < 0.05). Kaplan-Meier analysis showed the negative correlation between BTG1 mRNA expression and overall survival rate of all cancer patients (P < 0.05). BTG1 overexpression weakened adhesion and strengthened migration and invasion of HCT-15 cells (P < 0.05). There was E-cadherin hypoexpression, N-cadherin and MMP-9 hyperexpression, Zeb1 and Vimentin mRNA overexpression, a high expression of CEA mRNA and protein, and a strong secretion of CEA in BTG1 transfectants, compared with the control or mock. It was suggested that BTG1 expression might promote invasion and metastasis by decreasing adhesion, and inducing epithelial-mesenchymal transition.

7.
Front Oncol ; 10: 600322, 2020.
Article in English | MEDLINE | ID: mdl-33425757

ABSTRACT

Dkk3 has been discovered during comparison of immortalized and parental cells. Its expression has been shown to reduce colony formation and induce apoptosis of cancer cells, acting as a tumor suppressor. Herein, we demonstrate that Dkk3 overexpression or protein treatment may inhibit colorectal cancer cell proliferation, migration, and invasion and that they may promote apoptosis and G2 phase arrest with hypoexpression of Bcl-2, cdc25B, cdc25c, N-cadherin, slug, and twist and hyperexpression of Bax and E-cadherin. This effect is consistent with that of recombinant Dkk3 exposure and blocked with anti-Dkk3 antibody. Dkk3 deletion in intestinal cells was not associated with the emergence of epithelial lesions; however, adenoma emerged after sodium desoxycholate treatment. At both mRNA and protein levels, Dkk3 expression was higher in normal than in cancer tissues (p<0.05). Dkk3 mRNA expression was negatively associated with its promoter methylation, growth pattern, differentiation, and favorable prognosis in the patients with colorectal cancer (p<0.05). Dkk3-related signal pathways in colorectal cancer included those of cellular adhesion and migration, melanogenesis, chemokine, Hedgehog, JAK-STAT, TOLL-like receptor, TGF-ß, MAPK, and calcium signaling (p<0.05). These findings indicate that Dkk3 expression levels can help assess cancer aggressiveness and patient prognosis. It might also suppress aggressive phenotypes and tumorigenesis as a molecular target in gene therapy.

8.
Front Oncol ; 10: 613679, 2020.
Article in English | MEDLINE | ID: mdl-33425768

ABSTRACT

Beclin 1 is encoded by Becn1, and plays a role in tumorigenesis, neurodegeneration, apoptosis and autophagy. Here, the aggressive phenotypes and relevant proteins were examined after Beclin 1 expression was altered in gastric cancer cells. We also observed the effects of Beclin 1 on gastric carcinogenesis using Becn1 knockout mice. Finally, clinicopathological significances of Beclin 1 expression were analyzed using meta- and bioinformatics analyses. Becn1 overexpression was found to inhibit proliferation, glucose metabolism, migration and invasion of gastric cancer cells, whereas its knockdown caused the opposite effects. Beclin 1 suppressed the tumor growth by decreasing proliferation and increasing apoptosis. The heterozygous abrogation of Becn1 in gastric pit, parietal and chief cells could not cause any epithelial lesion. Beclin 1-mediated chemoresistance was closely linked to the autophagy, Bax underexpression, and the overexpression of Bcl-2, LRP1, MDR1, and ING5. Bioinformatics analysis showed higher Becn1 mRNA expression in intestinal- than diffuse-type carcinomas (P<0.05), and in male than female gastric cancer patients (P<0.05). Becn1 hyperexpression was positively associated with both overall and progression-free survival rates of the cancer patients (P<0.05). Meta-analysis showed that down-regulated Beclin 1 expression in gastric cancer was positively with lymph node metastasis, TNM staging, dedifferentiation and poor prognosis (P<0.05). Becn1-related signal pathways in gastric cancer included prostate, lung, renal, colorectal, endometrial and thyroid cancers, glioma, and leukemia, the metabolism of amino acid, lipid and sugar, and some signal pathways of insulin, MAPK, TRL, VEGF, JAK-STAT, chemokine, p53, lysosome, peroxidome and ubiquitin-mediated protein degradation (P<0.05). These suggested that Beclin 1 might be considered as a potential marker of gastric carcinogenesis, aggressiveness and prognostic prediction, and as a target of gene therapy in gastric cancer.

9.
Cancer Med ; 7(9): 4554-4569, 2018 09.
Article in English | MEDLINE | ID: mdl-30091530

ABSTRACT

Neuroblastoma is the most common extracranial solid neuroendocrine cancer and is one of the leading causes of death in children. To improve clinical outcomes and prognosis, discovering new promising drugs and targeted medicine is essential. We found that applying Suberoylanilide hydroxamic acid (SAHA; Vorinostat, a histone deacetylase inhibitor) and MG132 (a proteasome inhibitor) to SH-SY5Y cells synergistically suppressed proliferation, glucose metabolism, migration, and invasion and induced apoptosis and cell cycle arrest. These effects occurred both concentration and time dependently and were associated with the effects observed with inhibitor of growth 5 (ING5) overexpression. SAHA and MG132 treatment increased the expression levels of ING5, PTEN, p53, Caspase-3, Bax, p21, and p27 but decreased the expression levels of 14-3-3, MMP-2, MMP-9, ADFP, Nanog, c-myc, CyclinD1, CyclinB1, and Cdc25c concentration dependently, similar to ING5. SAHA may downregulate miR-543 and miR-196-b expression to enhance the translation of ING5 protein, which promotes acetylation of histones H3 and H4. All three proteins (ING5 and acetylated histones H3 and H4) were recruited to the promoters of c-myc, Nanog, CyclinD1, p21, and p27 for complex formation, thereby regulating the mRNA expression of downstream genes. ING5 overexpression and SAHA and/or MG132 administration inhibited tumor growth in SH-SY5Y cells by suppressing proliferation and inducing apoptosis. The expression of acetylated histones H3 and ING5 may be closely linked to the tumor size of neuroblastomas. In summary, SAHA and/or MG132 can synergistically suppress the malignant phenotypes of neuroblastoma cells through the miRNA-ING5-histone acetylation axis and via proteasomal degradation, respectively. Therefore, the two drugs may serve as potential treatments for neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Vorinostat/pharmacology , Acetylation , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Cell Cycle/drug effects , Cell Line, Tumor , Disease Models, Animal , Energy Metabolism/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Immunohistochemistry , Leupeptins/pharmacology , Mice , MicroRNAs/genetics , Models, Biological , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
10.
Oncotarget ; 8(11): 18322-18336, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28407690

ABSTRACT

Here, we found that down-regulated expression of BTG3 might be positively correlated with colorectal carcinogenesis and its overexpression suppressed proliferation, glycolysis, mitochondrial respiration, cell cycle progression, migration, and invasion, and induced apoptosis, senescence and differentiation in SW480 and SW620 cells. After treated with cisplatin, MG132, paclitaxel and SAHA, BTG3 transfectants exhibited lower viability and higher apoptosis than the control in both time- and dose-dependent manners. BTG3 overexpression up- regulated the protein expression of Cyclin E, p16, p27, NF-κB, p38α/ß, XIAP, Bcl-2, ATG14 and p53, but down-regulated the mRNA expression of MRP1, BCRP, and mTOR in SW480 and SW620 cells. BTG3 overexpression inhibited tumor growth of SW620 cells by suppressing proliferation and inducing apoptosis. It was suggested that down-regulated BTG3 expression might be considered as a marker for colorectal carcinogenesis. BTG3 overexpression might reverse the aggressive phenotypes and be employed as a potential target for gene therapy of colorectal cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/metabolism , Genetic Therapy/methods , Nuclear Proteins/metabolism , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Humans , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Phenotype , Signal Transduction , Transfection
11.
Oncotarget ; 8(14): 23603-23612, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28223542

ABSTRACT

Down-regulated parafibromin is positively linked to the pathogenesis of parathyroid, lung, breast, ovarian, gastric and colorectal cancers. Here, we found that wild-type (WT) parafibromin overexpression suppressed proliferation, tumor growth, induced cell cycle arrest and apoptosis in colorectal cancer cells (p<0.05), but it was the converse for mutant-type (MT, mutation in nucleus localization sequence) parafibromin (p<0.05). Both WT and MT transfectants inhibited migration and invasion, and caused better differentiation (p<0.05) of cancer cells. WT parafibromin transfectants showed the overexpression of Cyclin B1, Cyclin D1, Cyclin E, p38, p53, and AIF in HCT-15 and HCT-116 cells, while MT parafibromin only up-regulated p38 expression. There was lower mRNA expression of bcl-2 in parafibromin transfectants than the control and mock, while higher expression of c-myc, Cyclin D1, mTOR, and Raptor. According to transcriptomic analysis, WT parafibromin suppressed PI3K-Akt and FoxO signaling pathways, while MT one promoted PI3K-Akt pathway, focal adhesion, and regulation of actin cytoskeleton. Parafibromin was less expressed in colorectal cancer than paired mucosa (p<0.05), and inversely correlated with its differentiation at both mRNA and protein levels (p<0.05). These findings indicated that WT parafibromin might reverse the aggressive phenotypes of colorectal cancer cells and be employed as a target for gene therapy. Down-regulated parafibromin expression might be closely linked to colorectal carcinogenesis and cancer differentiation.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Cytoplasm/genetics , Cytoplasm/metabolism , Female , Gene Expression Profiling/methods , Genetic Therapy , HCT116 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mutation , Transplantation, Heterologous , Tumor Suppressor Proteins/metabolism
12.
Oncotarget ; 7(51): 84155-84164, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27677078

ABSTRACT

Here, we collected the information of 17304 and 2014 inpatients with colorectal cancer (CRC) from general hospitals of China and Japan respectively, and analyzed microscopic and macroscopic aspects, even stratified by the age and gender. It was found that Chinese CRC patients showed younger prone, more rectal and ascending cancers, less sigmoid and transverse cancers, larger size, less invasion into lymphatic system or metastasis into lymph node, and poorer differentiation than Japanese ones (p < 0.05). TNM staging was employed as an independent factor for the prognosis of the CRC patients regardless of the country (p < 0.05). Female patients showed larger tumor size, easier invasion and metastasis into lymphatic system, and worse differentiation than males (p < 0.05). The younger patients displayed frequent invasion and metastasis into lymphatic system, and poor differentiation in comparison to elder ones (p < 0.05). These findings demonstrated that Japanese patients seemed to have more invasion and metastasis due to standard and precise operation and pathological diagnosis accuracy. Actually, Chinese patients had more aggressive pathological characteristics and a poorer prognosis. Therefore, it is essential to establish a routine screening methodology, a standard treatment system and postoperative diagnosis protocol for the prevention and therapeutics of Chinese CRC patients, especially for female and young patients.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Hospitals, General/statistics & numerical data , Inpatients/statistics & numerical data , Aged , Asian People/statistics & numerical data , China , Colorectal Neoplasms/ethnology , Female , Humans , Japan , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prognosis
13.
Oncotarget ; 7(41): 67425-67434, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27608841

ABSTRACT

Here, we retrospectively compared the differences in clinicopathological behaviors and prognosis of lung cancer from the First Affiliated Hospital (CMU1, n=513), Shengjing Hospital (CMUS, n=1021), Tumor Hospital (CMUT, n=5378) of China Medical University, the First Affiliated Hospital of Dalian (DMU, n=2251) and Jinzhou (JMU, n=630) Medical University, Takaoka Kouseiren Hospital (Takaoka, n=163) of Japan. Japanese lung cancer patients showed smaller tumor size, lower TNM staging, lower ratio of squamous cell carcinoma and higher ratio of small and large cell carcinomas than Chinese patients (p<0.05). Survival analysis showed that tumor size was employed as a prognostic factor for the Japanese and Chinese cancer patients (p<0.05). In DMU and CMUS, the ratios of female patients or adenocarcinoma were higher than other hospitals (p<0.05), while the patients from CMUT and CMU1 were younger than the others (p<0.05). The ratios of squamous cell carcinoma from CMUT, CMU1 and JMU were higher than the others, while it was the same for the ratio of large and small cell carcinoma in Takaoka and CMU1 (p<0.05). TNM staging was higher in CMUT than JMU and Takaoka (p<0.05). The female patients of lung cancer showed young prone, large tumor size, a high ratio of adenocarcinoma and advanced TNM staging in comparison to the counterpart (p<0.05). The younger patients of lung cancer displayed smaller tumor size, higher ratio of adenocarcinoma, lower TNM staging than the elder in Takaoka (p<0.05). There were more aggressive behaviors and shorter survival time for Chinese than Japanese lung cancer patients. The prevention of lung cancer should be strengthened by establishing a systematic and effective screening strategy, especially for the young and female patients.


Subject(s)
Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Adult , Age Distribution , Aged , Aged, 80 and over , Asian People , China/epidemiology , Female , Humans , Inpatients , Japan/epidemiology , Male , Middle Aged , Prognosis , Retrospective Studies , Sex Distribution
14.
J BUON ; 20(5): 1244-9, 2015.
Article in English | MEDLINE | ID: mdl-26537071

ABSTRACT

PURPOSE: To investigate the effects of metformin (Met) on the proliferation and apoptosis of a renal carcinoma cell line and study the underlying mechanisms. METHODS: Renal cell carcinoma (RCC) line 786-O was cultured with media containing different concentrations of Met. The proliferation and apoptosis of 786-O cells were detected by the MTT method and flow cytometry, respectively. The invasion of 786-O cells was detected by scratch test, and the expression of pro-apoptotic proteins was determined by Western blot assay. RESULTS: The proliferation rates of 786-O cells with Met concentrations of 10, 15, and 20 mM were decreased by 62.8, 61.7, and 65.1%, respectively, with no significant difference among these concentrations (p>0.05). Twenty-four hrs after the scratch assay, the mean migration index in the control group and Met treatment group was 51.6% +/- 5.9 and 28.1% +/- 4.3, and that after 48 hrs was 92.2% +/- 6.4 and 68.0% +/- 4.9, respectively (p<0.05). At low serum concentration, the percentage of apoptotic cells in the Met treatment group (17.6%) was significantly higher (p<0.05) than that in the control group (5.2%). Met (10 mM) treatment significantly increased the expression of Caspase-3 and Bax proteins in 786-O cells (p<0.05). CONCLUSIONS: Metformin may be a potential drug of choice in the treatment of metastatic RCC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Metformin/pharmacology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Kidney Neoplasms/pathology
15.
Tumour Biol ; 35(4): 3333-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24272204

ABSTRACT

The tumor suppressor gene Smad4 has been localized to chromosome 18q21.1 and is a member of the Smad family that mediates the transforming growth factor ß signaling pathway suppressing epithelial cell growth. However, variable expression of Smad4 messenger RNA (mRNA) has been reported, with a loss in some cancers and increased expression in others. The aim of the present study was to investigate the Smad4 mRNA expression in prostate cancer tissues and adjacent noncancerous tissues and its potential relevance to clinicopathological variables and prognosis. The expression change of Smad4 mRNA was detected by using real-time quantitative reverse transcriptase-polymerase chain reaction analysis. The data showed that the Smad4 mRNA expression level in prostate cancer tissues was significantly lower than those in noncancerous tissues. The results indicated that the low expression of Smad4 mRNA in prostate cancer was associated with lymph node metastasis, preoperative prostate-specific antigen (PSA), and Gleason score. Kaplan-Meier survival analysis showed that patients with high Smad4 mRNA expression have longer biochemical recurrence-free survival time compared to patients with low Smad4 mRNA expression. Multivariate analysis revealed that Smad4 mRNA expression was an independent predictor of biochemical recurrence-free survival. Our results emphasize that Smad4 mRNA can be used as a predictive biomarker.


Subject(s)
Prostatic Neoplasms/genetics , RNA, Messenger/analysis , Smad4 Protein/genetics , Adult , Aged , Biomarkers, Tumor , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Recurrence, Local , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology
16.
Chemosphere ; 93(6): 1189-93, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23948605

ABSTRACT

Bisphenol A (BPA), the important endocrine-disrupting chemical (EDC), has been reported to be able to induce various toxicity. The present study aims to understand the toxicity behavior of bisphenol A through evaluating the inhibition profile of bisphenol A towards UDP-glucuronosyltransferase (UGT) isoforms. In vitro recombinant UGTs-catalyzed 4-methylumbelliferone (4-MU) glucuronidation reaction was employed as probe reaction for all the tested UGT isoforms. The results showed that bisphenol A exerted stronger inhibition towards UGT2B isoforms than UGT1A isoforms. Furthermore, the inhibition kinetic type and parameters (K(i)) were determined for the inhibition of bisphenol A towards UGT2B4, 2B7, 2B15, and 2B17. Bisphenol A exhibited the competitive inhibition towards UGT2B4, and noncompetitive inhibition towards UGT2B7, 2B15 and 2B17. The inhibition kinetic parameters (K(i)) were calculated to be 1.1, 32.6, 5.6, and 19.9 µM for UGT2B4, 2B7, 2B15 and 2B17, respectively. In combination with the in vivo concentration of bisphenol A, the elevation of exposure dose was predicted to increase by 29.1%, 1%, 5.7%, and 1.6% for UGT2B4, 2B7, 2B15, and 2B17, indicating the high influence of bisphenol A towards the in vivo UGT2B isofroms-mediated metabolism of xenobiotics and endogenous substances. All these data provide the supporting information for deeper understanding of toxicology of bisphenol A.


Subject(s)
Benzhydryl Compounds/toxicity , Enzyme Inhibitors/toxicity , Glucuronosyltransferase/metabolism , Phenols/toxicity , Xenobiotics/toxicity , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...