Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Infect Chemother ; 54(1): 140-152, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35384425

ABSTRACT

BACKGROUND: The kinetics of neutralizing antibodies against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) play an important role in evaluating vaccine efficacy and durability, herd immunity, additional vaccination, and prediction models of immune protection against coronavirus disease 2019. MATERIALS AND METHODS: Serum collection times were 4 and 8 weeks after 1st inoculation of AZD1222 (AstraZeneca, Cambridge, UK), and 2 and 16 weeks after 2nd inoculation with 12-week dosing intervals. Neutralizing antibody (Nab) titers were measured indirectly using commercially available R-FIND SARS-CoV-2 Neutralizing Antibody ELISA Kit (SG Medical Inc., Seoul, Korea). Possible influences of gender, age, and adverse events on neutralizing antibody titer were also investigated. RESULTS: Nab titers (median inhibition %) started to decrease shortly after reaching peaks. This decrease was more pronounced in the elderly group (≥56 years) than in the young group (≤39 years) at 8 weeks (49.5% vs. 55.4%, P = 0.021) and 16 weeks (40.6% vs. 53.9%, P = 0.006) after the 1st and 2nd inoculation. And Nab titers were inversely correlated with age in the 8-week (r = -0.2091, P = 0.0284) and the 28-week group (r = -0.2811, P = 0.0029). Seropositive conversion of Nab reached 89.1% and 100% following 1st and 2nd inoculation. This 100% seropositivity was dropped sharply to 74.5% after 16 weeks. Compared to subjects without adverse events (51.8%), median inhibition was higher in subjects with one or more systemic adverse events (74.2%, P = 0.0203) or those with one or more local and systemic adverse events (77.1%, P = 0.0003). CONCLUSION: Nab induced by AZD1222 (AstraZeneca, UK) vaccination started to degrade shortly after the production period. Nab titers were lower in the elderly than in younger group during the degradation period. This seems to be because the degradation process of Nab is more pronounced in the elderly. This may explain why the frequency of breakthrough infections, disease severity, and mortality were higher in the elderly and may require revaccination to ensure robust immunity.

3.
Perit Dial Int ; 39(6): 502-508, 2019.
Article in English | MEDLINE | ID: mdl-31582468

ABSTRACT

Background:Peritoneal dialysis (PD) has become an increasingly important treatment modality for end-stage renal disease. However, application of PD in patients with liver cirrhosis (LC) and subsequent outcomes have not been thoroughly evaluated.Methods:A total of 1,366 patients (≥ 18 years old) who started PD at 4 tertiary referral centers between January 2000 and December 2015 were initially reviewed. Among them, 45 patients with LC were finally analyzed (LC-PD). Using the multivariate Cox hazard ratio (HR) model, outcomes such as technique failure, infection, and mortality in patients with LC-PD were compared with those in non-LC-PD patients (non-LC-PD) or patients with LC who received hemodialysis (LC-HD). All of the patients were selected by 1:1 matching of age, sex, catheter insertion date, and diabetes mellitus.Results:During the mean follow-up duration of 43 ± 35.8 months, 12 patients with LC-PD experienced technique failure, and this rate was similar to that of non-LC-PD patients. In evaluating infection episodes, the most common causes for peritonitis and exit-site infection were Escherichia coli (5.8%) and Staphylococcus aureus (19.3%), respectively; these event rates of LC-PD did not differ from those of non-LC-PD. The all-cause mortality rate of the LC-PD group was not different from that of the non-LC-PD and LC-HD groups.Conclusion:Dialysis outcomes such as technique failure, infection, and mortality are not affected by the presence of LC. Accordingly, PD therapy is a good option in patients with LC.


Subject(s)
Kidney Failure, Chronic/therapy , Liver Cirrhosis/complications , Peritoneal Dialysis/adverse effects , Peritonitis/etiology , Biopsy , Female , Follow-Up Studies , Humans , Kidney Failure, Chronic/complications , Liver Cirrhosis/diagnosis , Magnetic Resonance Imaging , Male , Middle Aged , Peritonitis/diagnosis , Prognosis , Retrospective Studies , Tomography, X-Ray Computed , Treatment Failure , Ultrasonography
4.
J Clin Endocrinol Metab ; 103(9): 3376-3385, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29947765

ABSTRACT

Context: Müllerian-inhibiting substance/anti-Müllerian hormone (MIS/AMH) is produced in the ovarian granulosa cells, and it is believed to inhibit ovarian folliculogenesis and steroidogenesis in women of reproductive age. Objective: To investigate the expression of MIS/AMH type II receptor (MISRII/AMHRII) that binds MIS/AMH in the ovaries of reproductive-age women; to identify the exact targets of MIS/AMH. Design: Laboratory study using human ovarian tissue. Setting: University hospital. Patients: Tissue samples from 25 patients who had undergone ovarian surgery. Interventions: The segregation of ovarian granulosa and theca cells by laser microdissection was followed by RT-PCR, analyzing MISRII/AMHRII mRNA expression. Afterward, in situ hybridization and immunohistochemistry were performed to determine the localization of MISRII/AMHRII mRNA and protein expression. Main Outcome Measures: MISRII/AMHRII mRNA expression by RT-PCR, in situ hybridization, and immunohistochemistry. Results: MISRII/AMHRII were expressed in granulosa and theca cells of preantral and antral follicles. The granulosa cells showed stronger MISRII/AMHRII expression than theca cells. MISRII/AMHRII mRNA staining of granulosa and theca cells in large antral follicles, early atretic follicles, and corpus luteum waned but were still detected weakly, showing higher expression in theca cells than in granulosa cells. However, MISRII/AMHRII protein in the granulosa layer of the atretic follicle and corpus luteum could not be assessed. Conclusions: As MISRII/AMHRII is expressed in both granulosa and theca cells, this indicates that MIS/AMH, produced in the granulosa cells, is active in the theca cells as well. MIS/AMH is most likely actively involved not only in the autocrine and endocrine processes but also in the paracrine processes involving theca cells.


Subject(s)
Anti-Mullerian Hormone/metabolism , Ovary/cytology , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Theca Cells/metabolism , Adult , Corpus Luteum/metabolism , Female , Granulosa Cells/metabolism , Humans , Ovarian Follicle/metabolism , Ovary/metabolism
5.
Gastric Cancer ; 21(6): 956-967, 2018 11.
Article in English | MEDLINE | ID: mdl-29704153

ABSTRACT

BACKGROUND: Gastrokine 1 (GKN1) plays important roles in maintaining mucosal homeostasis, and in regulating cell proliferation and differentiation. Here, we determined whether GKN1 is a potential theragnostic marker for gastric cancer. METHODS: We identified GKN1 binding proteins using the protein microarray assay and investigated whether GKN1 is one of the exosomal cargo proteins by western blot, immunoprecipitation, and immunofluorescent assays. Cell proliferation and apoptosis were analyzed by MTT, BrdU incorporation, flow cytometry, and western blot assays. We further validated the functional relevance of exosomal GKN1 in MKN1-injected xenograft mice. The possibility of serum GKN1 as a diagnostic marker for gastric cancer was determined by ELISA assay. RESULTS: In protein microarray assay, GKN1 binding to 27 exosomal proteins was clearly observed. GKN1 was expressed in exosomes derived from HFE-145 gastric epithelial cells by western blot and immunofluorescent assays, but not in exosomes from AGS and MKN1 gastric cancer cells. Exosomes carrying GKN1 inhibited cell proliferation and induced apoptosis in both AGS and MKN1 cells, and exosomes carrying GKN1-treated nude mice-bearing MKN1 xenograft tumors exhibited significantly reduced tumor volume and tumor weight. Silencing of clathrin markedly down-regulated the internalization of exosomal GKN1. Interestingly, serum GKN1 concentrations in patients with gastric cancer were significantly lower than those in healthy individuals and patients with colorectal and hepatocellular carcinomas. CONCLUSIONS: The GKN1 is secreted and internalized in the gastric epithelium by exosome-driven transfer, which inhibits gastric tumorigenesis and supports the clinical application of GKN1 protein in gastric cancer diagnosis and treatment.


Subject(s)
Peptide Hormones/metabolism , Stomach Neoplasms/blood , Animals , Biomarkers, Tumor/blood , Cell Line, Tumor , Cell Proliferation , Clathrin/metabolism , Enzyme-Linked Immunosorbent Assay , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice, Inbred BALB C , Molecular Targeted Therapy/methods , Peptide Hormones/blood , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
6.
Exp Mol Med ; 50(1): e420, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29303507

ABSTRACT

Recurrence and metastasis are major challenges in the management of hepatocellular carcinoma (HCC) patients after resection. To identify a metastasis-associated gene signature, we performed comparative gene expression analysis with recurrent HCC tissues from HCC patients who underwent partial or total hepatectomy and from non-metastatic primary HCC tissues. From this, we were able to identify genes associated with HCC recurrence. TCIRG1 (T-Cell Immune Regulator 1) was one of the aberrantly overexpressed genes in patients with recurrent HCC who had undergone total hepatectomy. The significant overexpression of TCIRG1 was confirmed using the Liver Hepatocellular Carcinoma dataset from The Cancer Genome Atlas. High expression of TCIRG1 was significantly associated with poor 5-year disease-free and recurrence-free survival of HCC patients. TCIRG1 knockdown suppressed tumor cell growth and proliferation in HCC cell lines; caused a significant increase in the proportion of cells in the G1/S phase of cell cycle; induced cell death; suppressed the metastatic potential of HCC cells by selectively regulating the epithelial-mesenchymal transition (EMT) regulatory proteins E-cadherin, N-cadherin, Fibronectin, Snail and Slug; and significantly attenuated the metastatic potential of ras-transformed NIH-3T3 cells in vitro and in vivo. These findings suggest that TCIRG1 functions as a metastatic enhancer by modulating growth, death and EMT in HCC cells. TCIRG1 could be a therapeutic target for the treatment of liver malignancy and metastasis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition/genetics , Liver Neoplasms/pathology , Vacuolar Proton-Translocating ATPases/genetics , Animals , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/surgery , Lung Neoplasms/secondary , Male , Mice, Nude , Neoplasm Recurrence, Local/genetics , Prognosis , Vacuolar Proton-Translocating ATPases/metabolism , Xenograft Model Antitumor Assays
7.
Hepatology ; 67(4): 1360-1377, 2018 04.
Article in English | MEDLINE | ID: mdl-29059470

ABSTRACT

An accurate tool enabling early diagnosis of hepatocellular carcinoma (HCC) is clinically important, given that early detection of HCC markedly improves survival. We aimed to investigate the molecular markers underlying early progression of HCC that can be detected in precancerous lesions. We designed a gene selection strategy to identify potential driver genes by integrative analysis of transcriptome and clinicopathological data of human multistage HCC tissues, including precancerous lesions, low- and high-grade dysplastic nodules. The gene selection process was guided by detecting the selected molecules in both HCC and precancerous lesion. Using various computational approaches, we selected 10 gene elements as a candidate and, through immunohistochemical staining, showed that barrier to autointegration factor 1 (BANF1), procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3 (PLOD3), and splicing factor 3b subunit 4 (SF3B4) are HCC decision markers with superior capability to diagnose early-stage HCC in a large cohort of HCC patients, as compared to the currently popular trio of HCC diagnostic markers: glypican 3, glutamine synthetase, and heat-shock protein 70. Targeted inactivation of BANF1, PLOD3, and SF3B4 inhibits in vitro and in vivo liver tumorigenesis by selectively modulating epithelial-mesenchymal transition and cell-cycle proteins. Treatment of nanoparticles containing small-interfering RNAs of the three genes suppressed liver tumor incidence as well as tumor growth rates in a spontaneous mouse HCC model. We also demonstrated that SF3B4 overexpression triggers SF3b complex to splice tumor suppressor KLF4 transcript to nonfunctional skipped exon transcripts. This contributes to malignant transformation and growth of hepatocyte through transcriptional inactivation of p27Kip1 and simultaneously activation of Slug genes. CONCLUSION: The findings suggest molecular markers of BANF1, PLOD3, and SF3B4 indicating early-stage HCC in precancerous lesion, and also suggest drivers for understanding the development of hepatocarcinogenesis. (Hepatology 2018;67:1360-1377).


Subject(s)
Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , RNA Splicing Factors/metabolism , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Immunohistochemistry , Kruppel-Like Factor 4 , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Mice , Rats , Tissue Array Analysis/methods
8.
J Pathol ; 244(1): 107-119, 2018 01.
Article in English | MEDLINE | ID: mdl-28991363

ABSTRACT

MicroRNAs (miRNAs) engage in complex interactions with the machinery that controls the transcriptome and concurrently target multiple mRNAs. Here, we demonstrate that microRNA-495-3p (miR-495-3p) functions as a potent tumor suppressor by governing ten oncogenic epigenetic modifiers (EMs) in gastric carcinogenesis. From the large cohort transcriptome datasets of gastric cancer (GC) patients available from The Cancer Genome Atlas (TCGA) and the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO), we were able to recapitulate 15 EMs as significantly overexpressed in GC among the 51 EMs that were previously reported to be involved in cancer progression. Computational target prediction yielded miR-495-3p, which targets as many as ten of the 15 candidate oncogenic EMs. Ectopic expression of miRNA mimics in GC cells caused miR-495-3p to suppress ten EMs, and inhibited tumor cell growth and proliferation via caspase-dependent and caspase-independent cell death processing. In addition, in vitro metastasis assays showed that miR-495-3p plays a role in the metastatic behavior of GC cells by regulating SLUG, vimentin, and N-cadherin. Furthermore, treatment of GC cells with 5-aza-2'-deoxcytidine restored miR-495-3p expression; sequence analysis revealed hypermethylation of the miR-495-3p promoter region in GC cells. A negative regulatory loop is proposed, whereby DNMT1, among ten oncogenic EMs, regulates miR-495-3p expression via hypermethylation of the miR-495-3p promoter. Our findings suggest that the functional loss or suppression of miR-495-3p triggers overexpression of multiple oncogenic EMs, and thereby contributes to malignant transformation and growth of gastric epithelial cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Epigenomics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Stomach Neoplasms/pathology , Animals , Cadherins/metabolism , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , DNA Methylation , Genes, Reporter , Genes, Tumor Suppressor , Humans , Male , Mice , Stomach/pathology , Stomach Neoplasms/genetics
9.
Health Care Manage Rev ; 43(3): 182-192, 2018.
Article in English | MEDLINE | ID: mdl-28005677

ABSTRACT

BACKGROUND: Despite the increasing interest for Lean and Six Sigma implementations in hospitals, there has been little empirical evidence that goes beyond descriptive case studies to address the current status and the effectiveness of the implementations. PURPOSE: The aim of this study was to explore existing patterns of Lean and Six Sigma implementation in U.S. hospitals and compare the performance of the different patterns. METHODOLOGY/APPROACH: We collected data from 215 U.S. hospitals via a survey that includes measurement items developed from related literature. Using the cross-sectional data, we conducted a cluster analysis, followed by t tests, chi-square tests, and regression analyses for cluster verification. RESULTS: The cluster analysis identifies two clusters, a Moderate Six Sigma group and a Lean Six Sigma group. Results show that the Lean Six Sigma group outperforms the Moderate Six Sigma group across many performance dimensions: responsiveness capability, patient safety, and possibly cost saving. In addition, the Lean Six Sigma group tends to be composed of larger, private teaching hospitals located in more urban areas, and they employ more resources for quality improvement. CONCLUSION: Our research contributes to the quality management literature by supporting the possible complementary relationship between Lean and Six Sigma in hospitals. PRACTICE IMPLICATIONS: Our study encourages practitioners and managers to pay more attention to Lean implementation. Although Lean seems to be conducted in a limited fashion in many hospitals, it should be expanded and combined with Six Sigma for better results.


Subject(s)
Efficiency, Organizational , Implementation Science , Quality Improvement/organization & administration , Total Quality Management/organization & administration , Cross-Sectional Studies , Hospitals/statistics & numerical data , Humans , Patient Safety/statistics & numerical data , Surveys and Questionnaires , Total Quality Management/economics , United States
10.
Sci Rep ; 7(1): 2807, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28584243

ABSTRACT

NKX6.3 plays an important role in gastric epithelial differentiation and also acts as a gastric tumor suppressor. The specific aim of this study was to determine whether NKX6.3 contributes to gastric mucosal barrier function by regulating reactive oxygen species (ROS) production. NKX6.3 reduced ROS production and regulated expression of anti-oxidant genes, including Hace1. In addition, NKX6.3 reduced DNMT1 expression and activity by down-regulating NF-kB family gene transcription. Silencing of Hace1 recovered ROS production, whereas knock-down of DNMT1 and NF-kB reduced ROS production and induced Hace1 expression by hypomethylating its promoter region. In addition, NKX6.3 inhibited CagA effects on cell growth, ROS production, and NF-kB and DNMT1 activity. In gastric mucosae and cancers, NKX6.3 and Hace1 expression was significantly reduced. The NKX6.3 expression was positively correlated with Hace1 and Nrf2 genes, but negatively correlated with DNMT1. Hypermethylation of Hace1 gene was observed only in gastric mucosae with H. pylori, atrophy and intestinal metaplasia. Thus, these results suggest that NKX6.3 inhibits ROS production by inducing the expression of Hace1 via down-regulating NF-kB and DNMT1 activity in gastric epithelial cells.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Gastric Mucosa/metabolism , Homeodomain Proteins/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Enzyme Activation , Gene Expression Regulation , Humans , Oxidative Stress/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Gastric Cancer ; 20(5): 772-783, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28150071

ABSTRACT

BACKGROUND: GKN2 and TFF1 form a heterodimer that is only generated in the mucus-secreting cells of the normal stomach. The formation of this heterodimer is frequently disrupted in gastric cancer. However, the precise roles of GKN2 alone and in the heterodimer with TFF1 as well as the contributions of GKN2 and the heterodimer to gastric carcinogenesis are poorly understood. METHODS: Cell viability, proliferation, and apoptosis were analyzed in AGS, MKN1, MKN28, and MKN45 gastric cancer cells transfected with GKN2 and/or TFF1 using MTT, BrdU incorporation, and apoptosis assays, respectively. In addition, cell viability was examined in HFE-145 non-neoplastic gastric epithelial cells after GKN2 and/or TFF1 silencing. Furthermore, the cell cycle and the expression of cell cycle and apoptosis related proteins were assessed. The interaction between GKN2 and TFF1 was confirmed by co-immunoprecipitation. Immunohistochemistry was employed to explore TFF1 expression in 169 gastric cancer tissues. RESULTS: Co-transfection with GKN2 and TFF1 significantly inhibited cell viability and proliferation by inducing G1/S cell cycle arrest and suppressing positive cell cycle regulators. Simultaneous knockdown of GKN2 and TFF1 in HFE-145 cells resulted in markedly increased cell viability. Moreover, the interaction of GKN2 and TFF1 promoted cell death by enhancing caspase-3/7 activity and upregulating pro-apoptotic proteins. At the mRNA level, GKN2 and TFF1 were found to be positively correlated in non-tumor and tumor samples. Immunohistochemistry revealed loss of TFF1 expression in 128 (75.73%) of 169 gastric cancers. There was a borderline-significant association between GKN2 and TFF1 protein expression in gastric cancers (P = 0.0598). CONCLUSION: Collectively, our data demonstrated that the interaction between GKN2 and TFF1 can have synergistic antiproliferative and pro-apoptotic effects on gastric cancer.


Subject(s)
Apoptosis/genetics , Carrier Proteins/genetics , Stomach Neoplasms/genetics , Trefoil Factor-1/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques , Humans , Immunoprecipitation , RNA, Messenger/metabolism , Stomach Neoplasms/pathology , Transfection
12.
Gastric Cancer ; 20(2): 274-285, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27250838

ABSTRACT

BACKGROUND: We investigated whether GKN1, a gastric tumor suppressor, contributes to the progression of gastric cancer by regulating RhoA expression. METHODS: We analyzed the expression of GKN1, RhoA, miR-185, and miR-34a in 35 gastric cancer tissues, and compared their expression with T category and TNM stage. Cell migration and invasion, as well as the expression of epithelial-to-mesenchymal transition (EMT)-related proteins, were assessed in GKN1- and RhoA small interfering RNA (siRhoA)-transfected and recombinant-GKN1-treated AGS and MKN1 gastric cancer cells. RESULTS: Expression of RhoA protein and messenger RNA (mRNA) was increased in 15 (42.9 %) and 17 (48.6 %) of 35 gastric cancer tissues respectively, and was associated with higher T category and TNM stage. GKN1 expression was significantly decreased in 27 gastric cancers (77.1 %) with a higher T category, and was inversely correlated with RhoA mRNA expression. In AGS and MKN1 cells, GKN1 expression increased miR-185 and miR-34a expression and reduced RhoA mRNA and protein expression. A positive relationship between GKN1 and miR-34a and miR-185 expression and an inverse relationship between miR-34a and RhoA expression were observed in gastric cancer tissues. Cell migration and invasiveness were markedly decreased in GKN1- and siRhoA-transfected cells. GKN1 expression and silencing of RhoA decreased the expression of the proteins Snail, Slug, and vimentin. Furthermore, miR-185 and miR-34a silencing in MKN1 cells transfected with GKN1 stimulated cell migration and invasion, and increased the expression of EMT-related proteins. CONCLUSION: Our data suggest that GKN1 may inhibit gastric cancer cell migration and invasion by downregulating RhoA expression in a miR-185- and miR-34a-dependent manner.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , MicroRNAs/genetics , Peptide Hormones/pharmacology , Stomach Neoplasms/pathology , rhoA GTP-Binding Protein/antagonists & inhibitors , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Cell Adhesion , Cell Proliferation , Down-Regulation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
13.
EBioMedicine ; 9: 97-109, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27333045

ABSTRACT

Despite ongoing research and recent progress, the prognosis for patients with advanced gastric cancer remains poor. Wnt/ß-catenin and Rho-GTPase signaling pathways are known to play essential roles in malignant transformation and progression of various tumors, including gastric cancer. Here, we identify that NKX6 transcription factor, locus 3 (NKX6.3) binds directly to specific promoter regions of Wnt/ß-catenin and Rho-GTPase pathway-related genes, resulting in inhibition of cancer cell migration and invasion. Additionally, we find that the expression level of NKX6.3 is involved in regulation of gastric cancer progression and expression of Wnt/ß-catenin and Rho-GTPase pathway-related genes in clinical samples. These results suggest that NKX6.3 prevents EMT and cell migration, implying that NKX6.3 inactivation might be one of the key mechanisms of gastric cancer cell invasion and metastasis.


Subject(s)
Homeodomain Proteins/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement , Disease Progression , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Kaplan-Meier Estimate , Microscopy, Fluorescence , Neoplasm Staging , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Transcription Factors/chemistry , Transcription Factors/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism , rho GTP-Binding Proteins/genetics
14.
Chem Commun (Camb) ; 52(22): 4132-5, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26857551

ABSTRACT

Overproduction of hydrogen peroxide is involved in the pathogenesis of inflammatory diseases such as cancer and arthritis. To image hydrogen peroxide via chemiluminescence resonance energy transfer in the near-infrared wavelength range, we prepared quantum dots functionalized with a luminol derivative.


Subject(s)
Luminol/chemistry , Nanoparticles , Quantum Dots , Animals , Energy Transfer , Luminescence , Mice
15.
Oncotarget ; 7(10): 11412-23, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26863632

ABSTRACT

H2A.Z is a highly conserved H2A variant, and two distinct H2A.Z isoforms, H2A.Z.1 and H2A.Z.2, have been identified as products of two non-allelic genes, H2AFZ and H2AFV. H2A.Z has been reported to be overexpressed in breast, prostate and bladder cancers, but most studies did not clearly distinguish between isoforms. One recent study reported a unique role for the H2A.Z isoform H2A.Z.2 as a driver of malignant melanoma. Here we first report that H2A.Z.1 plays a pivotal role in the liver tumorigenesis by selectively regulating key molecules in cell cycle and epithelial-mesenchymal transition (EMT). H2AFZ expression was significantly overexpressed in a large cohort of hepatocellular carcinoma (HCC) patients, and high expression of H2AFZ was significantly associated with their poor prognosis. H2A.Z.1 overexpression was demonstrated in a subset of human HCC and cell lines. H2A.Z.1 knockdown suppressed HCC cell growth by transcriptional deregulation of cell cycle proteins and caused apoptotic cell death of HCC cells. We also observed that H2A.Z.1 knockdown reduced the metastatic potential of HCC cells by selectively modulating epithelial-mesenchymal transition regulatory proteins such as E-cadherin and fibronectin. In addition, H2A.Z.1 knockdown reduced the in vivo tumor growth rate in a mouse xenograft model. In conclusion, our findings suggest the oncogenic potential of H2A.Z.1 in liver tumorigenesis and that it plays established role in accelerating cell cycle transition and EMT during hepatocarcinogenesis. This makes H2A.Z.1 a promising target in liver cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Histones/genetics , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Hep G2 Cells , Heterografts , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Transfection
16.
Gastric Cancer ; 19(2): 381-391, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25752269

ABSTRACT

BACKGROUND: Gastrokine 1 (GKN1) acts as a gastric tumor suppressor. Here, we investigated whether GKN1 contributes to the maintenance of gastric mucosal homeostasis by regulating gastrin-induced gastric epithelial cell growth. METHODS: We assessed the effects of gastrin and GKN1 on cell proliferation in stable AGS(GKN1) and MKN1(GKN1) gastric cancer cell lines and HFE-145 nonneoplastic epithelial cells. Cell viability and proliferation were analyzed by MTT and BrdU incorporation assays, respectively. Cell cycle and expression of growth factor receptors were examined by flow cytometry and Western blot analyses. RESULTS: Gastrin treatment stimulated a significant time-dependent increase in cell viability and proliferation in AGS(mock) and MKN1(mock), but not in HFE-145, AGS(GKN1), and MKN1(GKN1), cells, which stably expressed GKN1. Additionally, gastrin markedly increased the S-phase cell population, whereas GKN1 significantly inhibited the effect of gastrin by regulating the expression of G1/S cell-cycle regulators. Furthermore, gastrin induced activation of the NF-kB and ß-catenin signaling pathways and increased the expression of CCKBR, EGFR, and c-Met in AGS and MKN1 cells. However, GKN1 completely suppressed these effects of gastrin via downregulation of gastrin/CCKBR/growth factor receptor expression. Moreover, GKN1 reduced gastrin and CCKBR mRNA expression in AGS and MKN1 cells, and there was an inverse correlation between GKN1 and gastrin, as well as between GKN1 and CCKBR mRNA expression in noncancerous gastric mucosae. CONCLUSION: These data suggest that GKN1 may contribute to the maintenance of gastric epithelial homeostasis and inhibit gastric carcinogenesis by downregulating the gastrin-CCKBR signaling pathway.


Subject(s)
Gastric Mucosa/metabolism , Gastrins/metabolism , Peptide Hormones/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gastric Mucosa/cytology , Gastric Mucosa/pathology , Gastrins/genetics , Gastrins/pharmacology , Gene Expression Regulation , Humans , NF-kappa B/metabolism , Peptide Hormones/metabolism , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Receptors, Growth Factor/metabolism , Stomach Neoplasms/pathology
17.
World J Gastroenterol ; 21(31): 9328-36, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26309358

ABSTRACT

AIM: To investigate the functional consequences of rs2736100 polymorphism in telomere length and examine its link to gastric cancer risk. METHODS: Telomere length and human telomerase reverse transcriptase (hTERT) mRNA expression were measured in 35 gastric cancer tissues and 5 cell lines and correlated to rs2736100 polymorphism. The relationship between rs2736100 polymorphism and the risk of gastric cancer were examined in 243 gastric cancer patients and 246 healthy individuals. RESULTS: The rs2736100 A allele carrier is closely associated with reduced hTERT mRNA expression and shortened telomere length in gastric cancer tissue and cell lines. When gastric cancers were stratified by histological subtype, telomere length and hTERT mRNA levels were significantly increased in those with the C/C genotype in intestinal-type gastric cancer, but not in diffuse-type gastric cancer. Interestingly, there was no significant difference in the genotype and allele frequencies of the rs2736100 polymorphism between the patients with gastric cancer and healthy controls. CONCLUSION: The rs2736100 polymorphism of the hTERT gene is involved in the regulation of hTERT expression and telomere length, but not in the risk of gastric cancer.


Subject(s)
Adenocarcinoma/genetics , Polymorphism, Single Nucleotide , Stomach Neoplasms/genetics , Telomerase/genetics , Telomere Homeostasis , Telomere/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Line, Tumor , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Phenotype , Risk Factors , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Telomerase/metabolism , Telomere/genetics , Young Adult
18.
Oncotarget ; 6(29): 28425-39, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26314965

ABSTRACT

NKX6.3 transcription factor is known to be an important regulator in gastric mucosal epithelial differentiation. The present study aimed to investigate whether NKX6.3 acts as an essential tumor suppressor in gastric carcinogenesis. Absent or reduced protein expression and decreased DNA copy number and mRNA transcript of the NKX6.3 gene were frequently observed in gastric cancers. Overexpression of NKX6.3 in AGSNKX6.3 and MKN1NKX6.3 cells markedly arrested cell proliferation by inhibiting cell cycle progression and induced apoptosis through both death receptor- and mitochondrial-pathways. In addition, stable NKX6.3 transfectants increased the expression of gastric differentiation markers, including SOX2 and Muc5ac, and decreased the expression of intestinal differentiation markers, CDX2 and Muc2. In ChIP-cloning and sequencing analyses, NKX6.3 coordinated a repertoire of target genes, some of which are clearly associated with cell cycle, differentiation and death. In particular, NKX6.3 transcriptional factor was found to bind specifically to the upstream sequences of GKN1, a gastric-specific tumor suppressor, and dramatically increase expression of the latter. Furthermore, there was a positive correlation between NKX6.3 and GKN1 expression in non-cancerous gastric mucosae. Thus, these data suggest that NKX6.3 may control the fate of gastric mucosal cells and function as a gastric tumor suppressor.


Subject(s)
Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Gastric Mucosa/metabolism , Homeodomain Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Immunoblotting , Mutation , Peptide Hormones/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism
19.
Oncotarget ; 6(15): 12970-83, 2015 May 30.
Article in English | MEDLINE | ID: mdl-26025919

ABSTRACT

Blood transcriptome reflects the status of diseases, and characteristic molecular signature provides a novel window on gene expression preceding acute coronary events. We aim to determine blood transcriptome-based molecular signature of acute coronary syndrome (ACS), and to identify novel serum biomarkers for early stage ST-segment-elevation myocardial infarction (STEMI). We obtained peripheral blood from the patients with ACS who visited emergency department within 4 hours after the onset of chest pain: STEMI (n = 10), Non-ST-segment-elevation MI (NSTEMI, n = 10) and unstable angina (UA, n = 11). Blood transcriptome scans revealed that a characteristic gene expression change exists in STEMI, resulting in 531 outlier genes as STEMI molecular signature (Welch's t test, P < 0.05). Another analysis with a set of blood samples of patients with STEMI (n = 7) before and 7 days after the primary percutaneous coronary intervention (n = 7) and normal control (n = 10) evidenced that STEMI molecular signature directly reflects the onset of STEMI pathogenesis. From the two sets of transcriptome-based STEMI signatures, we identified 10 genes encoding transmembrane or secretory proteins that are highly expressed in STEMI. We validated blood protein expression levels of these 10 putative biomarkers in 40 STEMI and 32 healthy subjects by ELISA. Data suggested that PGLYRP1, IRAK3 and VNN3 are more specific and sensitive diagnostic biomarkers for STEMI than traditional CK-MB or troponin.Blood transcriptome scans of ACS evidenced early stage molecular markers for STEMI. Here, we report novel biomarkers to diagnose STEMI at emergency department in hospitals by a simple ELISA method.


Subject(s)
Acute Coronary Syndrome/blood , Myocardial Infarction/blood , Acute Coronary Syndrome/genetics , Acute Coronary Syndrome/physiopathology , Acute Coronary Syndrome/surgery , Biomarkers/blood , Case-Control Studies , Early Diagnosis , Female , Humans , Male , Middle Aged , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Percutaneous Coronary Intervention , RNA/blood , RNA/genetics , Transcriptome
20.
J Hepatol ; 63(2): 408-19, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25817558

ABSTRACT

BACKGROUND & AIMS: Most common reason behind changes in histone deacetylase (HDAC) function is its overexpression in cancer. However, among HDACs in liver cancer, HDAC6 is uniquely endowed with a tumor suppressor, but the mechanism underlying HDAC6 inactivation has yet to be uncovered. METHODS: Microarray profiling and target prediction programs were used to identify miRNAs targeting HDAC6. A series of inhibitors, activators and siRNAs was introduced to validate regulatory mechanisms for microRNA-221-3p (miR-221) governing HDAC6 in hepatocarcinogenesis. RESULTS: Comprehensive miRNA profiling analysis identified seven putative endogenous miRNAs that are significantly upregulated in hepatocellular carcinoma (HCC). While miR-221 was identified as a suppressor of HDAC6 by ectopic expression of miRNA mimics in Dicer knockdown cells, targeted-disruption of miR-221 repressed cancer cell growth through derepressing HDAC6 expression. Suppression of HDAC6 via miR-221 was induced by JNK/c-Jun signaling in liver cancer cells but not in normal hepatic cells. Additionally, cytokine-induced NF-κBp65 independently regulated miR-221, thereby suppressing HDAC6 expression in HCC cells. HCC tissues derived from chemical-induced rat and H-ras12V transgenic mice liver cancer models validated that JNK/c-Jun activation and NF-κBp65 nuclear translocation are essential for the transcription of miR-221 leading to repression of HDAC6 in HCC. CONCLUSIONS: Our findings suggest that the functional loss or suppression of the tumor suppressor HDAC6 is caused by induction of miR-221 through coordinated JNK/c-Jun- and NF-κB-signaling pathways during liver tumorigenesis, providing a novel target for the molecular treatment of liver malignancies.


Subject(s)
Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Liver Neoplasms, Experimental/genetics , MicroRNAs/genetics , RNA, Neoplasm/genetics , Animals , Disease Progression , Histone Deacetylase 6 , Histone Deacetylases/biosynthesis , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , Polymerase Chain Reaction , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...