Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
J Gene Med ; 26(1): e3616, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38049938

ABSTRACT

BACKGROUND: Upper tract urothelial carcinoma (UTUC) is a rare tumor with extraordinarily different features between Eastern and Western countries. Vascular endothelial growth factor-A (VEGFA) was originally identified as a secreted signaling protein and regulator of vascular development and cancer progression. In this study, we aimed to elucidate the molecular mechanisms underlying the regulation of VEGFA by microRNA in UTUC. METHODS: VEGFA expression was evaluated by immunohistochemistry in 140 human UTUC tissue samples. Next, we assessed the regulatory relationship between VEGFA and miR-299-3p by real-time PCR, western blotting, ELISA and dual-luciferase reporter assays using two UTUC cell lines. The role of miR-299-3p/VEGFA in cell proliferation, motility, invasion, and tube formation was analyzed in vitro. RESULTS: High VEGFA expression was significantly associated with tumor stage, grade, distant metastasis and cancer-related death and correlated with poor progression-free and cancer-specific survival. VEGFA knockdown repressed proliferation, migration, invasion and angiogenesis in UTUC cell lines. miR-299-3p significantly reduced VEGFA protein expression and miR-299-3p overexpression inhibited VEGFA mRNA and protein expression by directly targeting its 3'-UTR. Functional studies indicated that VEGFA overexpression reversed the miR-299-3p-mediated suppression of tumor cell proliferation, migration, invasion and angiogenesis. In addition, miR-299-3p/VEGFA suppressed cellular functions in UTUC by modulating the expression of P18 and cyclin E2. CONCLUSIONS: Our findings suggest that miR-299-3p possibly suppresses UTUC cell proliferation, motility, invasion and angiogenesis via VEGFA. VEGFA may act as a prognostic predictor, and both VEGFA and miR-299-3p could be potential therapeutic targets for UTUC.


Subject(s)
Carcinoma, Transitional Cell , MicroRNAs , Urinary Bladder Neoplasms , Humans , Angiogenesis , Carcinoma, Transitional Cell/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
J Transl Med ; 20(1): 324, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864526

ABSTRACT

Kidney transplantation is a lifesaving option for patients with end-stage kidney disease. In Taiwan, urothelial carcinoma (UC) is the most common de novo cancer after kidney transplantation (KT). UC has a greater degree of molecular heterogeneity than do other solid tumors. Few studies have explored genomic alterations in UC after KT. We performed whole-exome sequencing to compare the genetic alterations in UC developed after kidney transplantation (UCKT) and in UC in patients on hemodialysis (UCHD). After mapping and variant calling, 18,733 and 11,093 variants were identified in patients with UCKT and UCHD, respectively. We excluded known single-nucleotide polymorphisms (SNPs) and retained genes that were annotated in the Catalogue of Somatic Mutations in Cancer (COSMIC), in the Integrative Onco Genomic cancer mutations browser (IntOGen), and in the Cancer Genome Atlas (TCGA) database of genes associated with bladder cancer. A total of 14 UCKT-specific genes with SNPs identified in more than two patients were included in further analyses. The single-base substitution (SBS) profile and signatures showed a relative high T > A pattern compared to COMSIC UC mutations. Ingenuity pathway analysis was used to explore the connections among these genes. GNAQ, IKZF1, and NTRK3 were identified as potentially involved in the signaling network of UCKT. The genetic analysis of posttransplant malignancies may elucidate a fundamental aspect of the molecular pathogenesis of UCKT.


Subject(s)
Carcinoma, Transitional Cell , Kidney Transplantation , Urinary Bladder Neoplasms , Humans , Mutation/genetics , Urinary Bladder Neoplasms/pathology , Exome Sequencing
3.
FASEB J ; 34(4): 5975-5992, 2020 04.
Article in English | MEDLINE | ID: mdl-32077148

ABSTRACT

ADP-ribosylation factor 6 (ARF6) is a well-studied protein that is involved in multiple biological functions including cell migration and invasion. The mechanism by which ARF6 regulates the migration and invasion of upper tract urothelial carcinoma (UTUC) is still unknown. MiR-145-5p is a tumor suppressor microRNA, which is downregulated in several cancer types. We aimed to elucidate the molecular mechanism underlying the regulation of ARF6 by miR-145-5p in UTUC. ARF6 expression was observed to be higher in UTUC tissues than paired adjacent normal tissues. A reverse correlation between ARF6 and miR-145-5p was found in UTUC tissues. MiR-145-5p inhibited ARF6 expression by directly targeting its 3'-UTR. The functional studies indicated that ARF6 expression reversed the miR-145-5p-reduced tumor cell migration and invasion. Notably, miR-145-5p reduced MMP2, N-cadherin, FAK and MMP7, and elevated E-cadherin protein levels in vitro; however, the above effects were reversed by ARF6. Further, the expression of epithelial-to-mesenchymal transition (EMT) markers and cell invasion was suppressed by knocking down MMP7 in UTUC cells. These findings suggest that miR-145-5p may suppress UTUC cell motility and invasion by targeting ARF6/MMP7 through EMT.


Subject(s)
ADP-Ribosylation Factors/metabolism , Biomarkers, Tumor/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Urologic Neoplasms/pathology , Urothelium/pathology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Aged , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Proliferation , Female , Humans , Male , Neoplasm Invasiveness , Prognosis , Retrospective Studies , Survival Rate , Tumor Cells, Cultured , Urologic Neoplasms/genetics , Urologic Neoplasms/metabolism , Urothelium/metabolism
4.
Pathol Int ; 71(7): 463-470, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33848380

ABSTRACT

Upper tract urothelial carcinoma (UTUC) is a rare tumor with an incidence that varies greatly between Eastern and Western countries. Transaldolase 1 (TALDO1) is a rate-limiting enzyme of the pentose phosphate pathway. In humans, aberrant TALDO1 activity has been implicated in various autoimmune diseases and malignancies; however, the function of TALDO1 in UTUC has not been previously investigated. Here we evaluated the clinical significance of TALDO1 expression in 115 paraffin-embedded tumor samples from patients with UTUC using immunohistochemistry. Our results demonstrated that there was an association between high TALDO1 expression and advanced stage (P = 0.011), tumor size (P = 0.005), tumor location (P = 0.047), distant metastases (P = 0.023), local recurrence (P = 0.002), and cancer death (P = 0.003). Using univariate and multivariate analyses, we found that chemotherapy was an independent factor for bladder recurrence-free survival. Late stage (III/IV) and high TALDO1 expression were independent prognostic factors for progression-free and cancer-specific survival. In summary, increased TALDO1 expression in UTUC was significantly correlated with late stage, tumor size, tumor location, distant metastases, local recurrence, and cancer death. Therefore, high TALDO1 expression could be a predictor of poor survival in patients with UTUC. Further studies are necessary to investigate the role of TALDO1 in UTUC development.


Subject(s)
Prognosis , Transaldolase/metabolism , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/pathology , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Recurrence, Local , Retrospective Studies
5.
Int J Med Sci ; 18(5): 1179-1184, 2021.
Article in English | MEDLINE | ID: mdl-33526978

ABSTRACT

The introduction of mammalian target of rapamycin inhibitors (mTORi) as immunosuppressive agents has changed the landscape of calcineurin inhibitor-based immunosuppressive regimens. However, the timing of mTORi conversion and its associated outcomes in kidney transplantation have conflicting results. This study investigated the effect of early or late mTORi post-transplant initiation on major transplant outcomes, including post-transplant malignancy, in kidney transplant recipients in our center. We enrolled 201 kidney transplant recipients with surviving function grafts of >3 months between 1983 and 2016. Patients were divided into three groups: early mTORi (initiated within 6 months of kidney transplantation), late mTORi, (mTORi initiation >6 months after kidney transplantation) and no mTORi. The mean creatinine at conversion was 1.46 ± 0.48 mg/dL and 1.30 ± 0.53 mg/dL for the early and late mTORi groups, respectively. During the study period, 10.5% of mTORi users and 19.2% of mTORi nonusers developed malignancy, mainly urothelial carcinoma. After adjustment for confounding factors, mTORi users were found to have a lower incidence of post-transplant malignancy than did nonusers (adjusted OR: 0.28, P = 0.04). No significant difference was observed between early and late mTORi users. Our results verified the potential advantages of mTORi usage in reducing cancer incidence after kidney transplantation. However, no significant result was found related to the timing of mTORi introduction. Future studies should include a longer observation period with a larger cohort.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Kidney Transplantation/adverse effects , Neoplasms/epidemiology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Calcineurin Inhibitors/adverse effects , Everolimus/administration & dosage , Female , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immunocompromised Host , Immunosuppressive Agents/administration & dosage , Incidence , Male , Middle Aged , Neoplasms/immunology , Neoplasms/prevention & control , Retrospective Studies , Risk Factors , Sirolimus/administration & dosage , Time-to-Treatment/statistics & numerical data , Treatment Outcome
6.
J Cell Biochem ; 118(6): 1563-1573, 2017 06.
Article in English | MEDLINE | ID: mdl-27918099

ABSTRACT

Transcription factor CCAAT/enhancer-binding protein delta (CEBPD) plays multiple roles in tumor progression. Studies have demonstrated that cisplatin (CDDP) induced CEBPD expression and had led to chemotherapeutic drug resistance. However, the underlying molecular mechanisms of CDDP-regulated CEBPD expression and its relevant roles in CDDP responses remain elusive. MicroRNAs (miRNAs) are small non-coding RNAs that negatively regulate gene expression in a sequence-specific manner. Abnormal miRNAs expression is associated with tumor progression. In current study, a large-scale PCR-based miRNA screening was performed to identify CEBPD-associated miRNAs in urothelial carcinoma cell line NTUB1. Eleven miRNAs were selected with more than twofold changes. MiR-193b-3p, a known tumor suppressor, down-regulated proto-oncogenes Cyclin D1, and ETS1 expression and led to cell cycle arrest, cell invasion, and migration inhibition. The expression of miR-193b-3p was associated with the DNA binding ability of CEBPD in CDDP response. CEBPD knocking-down approach provided a strong evidence of the positive correlation between CEBPD and miR-193b-3p. CDDP-induced CEBPD trans-activated miR-193b-3p expression and it directly targeted the 3'-UTR of Cyclin D1 and ETS1 mRNA, and silenced the protein expression. In addition, miR-193b-3p also inhibited cell migration activity, arrested cell at G1 phase, and sensitized NTUB1 to CDDP treatment. In conclusion, this study indicates that CEBPD exhibits an anti-tumorigenic function through transcriptionally activating miR-193b-3p expression upon CDDP treatment. This study provides a new direction for managing human urothelial carcinoma. J. Cell. Biochem. 118: 1563-1573, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/genetics , Carcinoma, Transitional Cell/genetics , Cyclin D1/genetics , Drug Resistance, Neoplasm , MicroRNAs/genetics , Proto-Oncogene Protein c-ets-1/genetics , Urinary Bladder Neoplasms/genetics , 3' Untranslated Regions , Carcinoma, Transitional Cell/drug therapy , Cell Cycle , Cell Line, Tumor , Cell Movement , Cisplatin/pharmacology , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Urinary Bladder Neoplasms/drug therapy
7.
World J Urol ; 34(6): 835-46, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26433386

ABSTRACT

PURPOSE: Bladder cancer (BC) is the most common malignancy in urinary system. The prognosis of metastatic BC is poor, but there remains no reliable marker to early detect metastasis. Dysregulated prenylated protein tyrosine phosphatases (PTPs) are commonly associated with cancer metastasis. From a published BC transcriptome, we identified that PTP IVA3 (PTP4A3) was the most significantly upregulated gene implicated in tumor progression among genes related to prenylated PTPs. We therefore analyzed PTP4A3 expression in our well-characterized cohort of BC. METHODS: By immunohistochemistry, PTP4A3 expression was determined using H-score. PTP4A3 expression of 295 BCs was compared with clinicopathological parameters, and the effect of PTP4A3 on cancer-specific survival (CSS) and metastasis-free survival (MFS) was also examined. Two independent sets of BCs were used to assess PTP4A3 protein and transcript expression in normal urothelium and different stage tumors. RESULTS: PTP4A3 overexpression was significantly associated with higher pT stage (P < 0.001), nodal metastasis (P < 0.001), vascular invasion (P < 0.001), and perineural invasion (P = 0.021). In multivariate analysis, PTP4A3 overexpression was an independent predictor for CSS (P < 0.001) and MFS (P = 0.007). Notably, the difference in CSS and MFS between high and low PTP4A3-expressing tumors was also significant in muscle-invasive BCs. PTP4A3 protein expression showed significant and stepwise increments from normal urothelium to noninvasive BC, invasive BC, and metastatic foci (P < 0.001). PTP4A3 transcript was also obviously upregulated in high-stage BC (P < 0.001). CONCLUSIONS: PTP4A3 may play a role in BC oncogenesis and is a predictive marker of metastasis. PTP4A3 overexpression represents an independent prognosticator for BC, suggesting its potential theranostic value.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Urinary Bladder Neoplasms/genetics , Aged , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Prognosis , Protein Tyrosine Phosphatases/biosynthesis , Retrospective Studies , Survival Rate , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
8.
Arch Toxicol ; 89(4): 591-605, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24888374

ABSTRACT

Recent studies have shown that docetaxel-based chemotherapy confers a survival benefit in patients with castration-resistant prostate cancer (PC). Also epidermal growth factor receptor (EGFR) was found to have multiple roles in prostatic tumorigenesis. However, the EGFR-mediated chemoresistance mechanism in human PC was not well delineated. In this study, we explored the mechanism of EGFR-mediated docetaxel resistance in PC. A series of stable docetaxel-resistant PC/DX sublines were established at our laboratory. The docetaxel IC50s of PC3 and PC/DX25 cells were 0.01 and 1.33 µM, respectively. Cellular resistance to docetaxel was significantly associated with increased EGFR and EGFR activation in PC/DX25. There was a dose-dependent increase in EGFR expression associated with the magnitude of docetaxel resistance. Expression of EGFR in PC/DX25 was higher than that in PC3, RWPE-1 and LNCaP cells. Similar results were also found in human PC tissues by immunohistochemical staining. We showed that docetaxel sensitivity can be stored in PC/DX25 cells by knockdown and inactivation of EGFR expression through EGFR siRNA and specific inhibitors, respectively. Contrarily, overexpression of EGFR or recombinant EGF protein treatment could rescue PC3 cells from docetaxel-mediated cytotoxicity. Gefitninb (ZD1839) significantly inhibited the growth of PC/DX25 cells by MTT in vitro and on xenografted nude mice in vivo. Moreover, EGFR-mediated docetaxel resistance occurred through the Akt-dependent ABCB1 expression in PC cells. These findings demonstrated EGFR played an important role in docetaxel-resistant PC and EGFR inhibition may enhance the therapeutic efficacy of docetaxel-based treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/therapeutic use , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Male , Mice, Inbred BALB C , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Taxoids/administration & dosage , Xenograft Model Antitumor Assays
9.
Biol Pharm Bull ; 37(7): 1191-8, 2014.
Article in English | MEDLINE | ID: mdl-24989010

ABSTRACT

In an effort to develop potent cyclooxygenase-1 (COX-1) inhibitors used as anticancer agent, a series of 2',5'-dimethoxychalcones was screened to evaluate their antiplatelet effect on human washed platelets suspension. Compound 2 exhibited potent inhibition of human washed platelet aggregation induced by collagen, significantly inhibited collagen- and arachidonic acid-induced thromboxane B2 release, and revealed inhibitory effect on COX-1 activity. Molecular docking studies showed that 1, 2, and 4 were bound in the active site of COX-1. These indicated that the antiplatelet effect of these compounds were mainly mediated through the suppression of COX-1 activity and reduced the thromboxane formation. To investigate the mechanistic action of COX-1 inhibitor enhanced the cytotoxic effect against human bladder cancer cells, NTUB1, we assessed the cytotoxic effect of 2 against NTUB1. Treatment of NTUB1 cells with various concentrations of 2 led to a concentration-dependent increase of cell death and decrease of reactive oxygen species levels. The flow-cytometric analysis showed that 2 induced a G1 phase cell cycle arrest but did not accompany an appreciable sub-G1 phase in NTUB1 cells. In addition, compound 2 increased p21 and p27 expressions and did not inhibit the expression of COX-1 in NTUB1 cells. Our results suggested that 2 enhanced cell growth inhibition or antiproliferative activity in NTUB1 cells through G1 arrest by COX-1 independent mechanism.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Chalcone/analogs & derivatives , Chalcone/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Chalcone/chemistry , Cyclooxygenase 1/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Platelet Aggregation Inhibitors/chemistry , Reactive Oxygen Species/metabolism
10.
Arch Pharm (Weinheim) ; 347(1): 42-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24243582

ABSTRACT

Ten new lantabetulic acid (1) derivatives 2-11 were synthesized and their cytotoxicities against human prostate cancer cells were evaluated. PC3 cells treated with 10 µM 8 exhibited the most potent G1 phase arrest. In addition, 10 µM 8 markedly decreased the levels of cyclin E and cdk2 and caused an increase in the p21 and p27 levels, while 20 µM 8 mainly led to cell death through the apoptotic pathway, which correlated with an increase in reactive oxygen species levels, decreased expression levels of Bcl-2 and caspase-8, the induction of mitochondrial changes, and decreased levels of cytochrome c in mitochondria. The dual action of 8 could provide a new approach for the development of chemotherapeutic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Prostatic Neoplasms/pathology , Triterpenes/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Molecular Structure , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Time Factors , Triterpenes/chemical synthesis
11.
Hu Li Za Zhi ; 61(4): 56-65, 2014 Aug.
Article in Zh | MEDLINE | ID: mdl-25116315

ABSTRACT

BACKGROUND: Little has been published in the literature regarding how patients self-evaluate their degree of readiness for hospital discharge. Furthermore, there is currently no self-evaluation tool available in Chinese able to assess the discharge readiness of patients. PURPOSE: This study was used to psychometrically test the Chinese version of the readiness for hospital discharge scale (RHDS_C). METHODS: This study used a cross-sectional design. Two samples were recruited in a two-stage process at two hospitals in Southern Taiwan. Two hundred and twenty-three patients with a diagnosis of either colorectal cancer or hepatic cancer were used to conduct an exploratory factor analysis (EFA) in the first stage of the study. Another 323 patients with a diagnosis of stroke were used conduct a confirmatory factor analysis (CFA). The instrument used was the Readiness for Hospital Discharge Scale (RHDS) developed by Weiss & Piacentine. RESULTS: RHDS_C consists of three subscales: personal status (4 items), coping ability (4 items), and expected support (4 items) adapted from the CFA. The assessed goodness-of-fit index (GFI = .92, AGFI = .88, NFI = .97) indicate the model fit the data well based upon the CFA. Criterion-related validity was supported by the correlation between the original RHDS and the RHDS_C (r = .96, p < .001). The Cronbach's alpha coefficients were .89 for the overall scale and .73, .90, and .89 for the 3 subscales, respectively. CONCLUSIONS / IMPLICATIONS FOR PRACTICE: This study confirms the validity of the RHDS_C and suggests this instrument is able to reliably assess the readiness of patients for discharge from the hospital. We recommend the scale be applied in the clinical setting to evaluate the discharge readiness of hospital patients.

12.
Kaohsiung J Med Sci ; 40(5): 445-455, 2024 May.
Article in English | MEDLINE | ID: mdl-38593276

ABSTRACT

Neurotrophic receptor tyrosine kinase 3 (NTRK3) has pleiotropic functions: it acts not only as an oncogene in breast and gastric cancers but also as a dependence receptor in tumor suppressor genes in colon cancer and neuroblastomas. However, the role of NTRK3 in upper tract urothelial carcinoma (UTUC) is not well documented. This study investigated the association between NTRK3 expression and outcomes in UTUC patients and validated the results in tests on UTUC cell lines. A total of 118 UTUC cancer tissue samples were examined to evaluate the expression of NTRK3. Survival curves were generated using Kaplan-Meier estimates, and Cox regression models were used for investigating survival outcomes. Higher NTRK3 expression was correlated with worse progression-free survival, cancer-specific survival, and overall survival. Moreover, the results of an Ingenuity Pathway Analysis suggested that NTRK3 may interact with the PI3K-AKT-mTOR signaling pathway to promote cancer. NTRK3 downregulation in BFTC909 cells through shRNA reduced cellular migration, invasion, and activity in the AKT-mTOR pathway. Furthermore, the overexpression of NTRK3 in UM-UC-14 cells promoted AKT-mTOR pathway activity, cellular migration, and cell invasion. From these observations, we concluded that NTRK3 may contribute to aggressive behaviors in UTUC by facilitating cell migration and invasion through its interaction with the AKT-mTOR pathway and the expression of NTRK3 is a potential predictor of clinical outcomes in cases of UTUC.


Subject(s)
Cell Movement , Receptor, trkC , Urologic Neoplasms , Female , Humans , Male , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Kaplan-Meier Estimate , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Receptor, trkC/metabolism , Receptor, trkC/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Urologic Neoplasms/genetics , Urologic Neoplasms/metabolism , Urologic Neoplasms/pathology
13.
Proc Natl Acad Sci U S A ; 107(37): 16131-6, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805509

ABSTRACT

Maintenance of genomic integrity is an essential cellular function. We previously reported that the transcription factor and tumor suppressor CCAAT/enhancer binding protein δ (C/EBPδ, CEBPD; also known as "NFIL-6ß") promotes genomic stability. However, the molecular mechanism was not known. Here, we show that C/EBPδ is a DNA damage-induced gene, which supports survival of mouse bone marrow cells, mouse embryo fibroblasts (MEF), human fibroblasts, and breast tumor cells in response to the DNA cross-linking agent mitomycin C (MMC). Using gene knockout, protein depletion, and overexpression studies, we found that C/EBPδ promotes monoubiquitination of the Fanconi anemia complementation group D2 protein (FANCD2), which is necessary for its function in replication-associated DNA repair. C/EBPδ interacts with FANCD2 and importin 4 (IPO4, also known as "Imp4" and "RanBP4") via separate domains, mediating FANCD2-IPO4 association and augmenting nuclear import of FANCD2, a prerequisite for its monoubiquitination. This study identifies a transcription-independent activity of C/EBPδ in the DNA damage response that may in part underlie its tumor suppressor function. Furthermore, we report a function of IPO4 and nuclear import in the Fanconi anemia pathway of DNA repair.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/metabolism , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/metabolism , Karyopherins/metabolism , Membrane Transport Proteins/metabolism , Active Transport, Cell Nucleus , Animals , CCAAT-Enhancer-Binding Protein-delta/deficiency , CCAAT-Enhancer-Binding Protein-delta/genetics , Cell Line , Cell Survival , Humans , Mice , Mice, Knockout , Protein Binding , Ubiquitination
14.
Proc Natl Acad Sci U S A ; 107(20): 9210-5, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20439707

ABSTRACT

The transcription factor CCAAT/enhancer binding protein delta (C/EBPdelta, CEBPD, NFIL-6beta) has tumor suppressor function; however, the molecular mechanism(s) by which C/EBPdelta exerts its effect are largely unknown. Here, we report that C/EBPdelta induces expression of the Cdc27 (APC3) subunit of the anaphase promoting complex/cyclosome (APC/C), which results in the polyubiquitination and degradation of the prooncogenic cell cycle regulator cyclin D1, and also down-regulates cyclin B1, Skp2, and Plk-1. In C/EBPdelta knockout mouse embryo fibroblasts (MEF) Cdc27 levels were reduced, whereas cyclin D1 levels were increased even in the presence of activated GSK-3beta. Silencing of C/EBPdelta, Cdc27, or the APC/C coactivator Cdh1 (FZR1) in MCF-10A breast epithelial cells increased cyclin D1 protein expression. Like C/EBPdelta, and in contrast to cyclin D1, Cdc27 was down-regulated in several breast cancer cell lines, suggesting that Cdc27 itself may be a tumor suppressor. Cyclin D1 is a known substrate of polyubiquitination complex SKP1/CUL1/F-box (SCF), and our studies show that Cdc27 directs cyclin D1 to alternative degradation by APC/C. These findings shed light on the role and regulation of APC/C, which is critical for most cellular processes.


Subject(s)
Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic/genetics , Proteasome Endopeptidase Complex/metabolism , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome , Blotting, Western , CCAAT-Enhancer-Binding Protein-delta/genetics , Cell Line, Tumor , Cyclin B1/metabolism , Gene Expression Regulation, Neoplastic/physiology , Immunoprecipitation , Mice , Mice, Knockout , Microscopy, Fluorescence , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , S-Phase Kinase-Associated Proteins/metabolism , Polo-Like Kinase 1
15.
Appl Immunohistochem Mol Morphol ; 31(2): 94-100, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36688483

ABSTRACT

Upper tract urothelial carcinoma (UTUC) is an aggressive malignancy with characteristics of high metastasis and poor prognosis. There are some particularly different features of UTUC between the Asian and Western countries. Double-strand break repair protein MRE11 is a component of the MRN complex that is involved in the DNA repair pathway. Emerging studies have focused on the role of MRE11 in human malignancies with conflicting results. We aimed to establish the relationship between MRE11 expression and the oncological outcome of UTUC. This study retrospectively reviewed 150 patients who underwent radical nephroureterectomy with pathologically confirmed UTUC. Pathologic slides were reviewed, and clinical parameters were collected. An immunohistochemical study was performed, and the cytoplasmic and nuclear-staining results of UTUC were recorded. The expression of MRE11 was analyzed to identify correlations with various clinicopathological parameters, metastasis-free survival, and cancer-specific survival (CSS). MRE11 expression was significantly correlated with patients with a high pathologic stage ( P =0.001), perineural invasion ( P =0.015), and tumor necrosis ( P =0.034). Upon univariate analysis, a high MRE11 expression was associated with poor metastasis-free survival ( P =0.014, 95% CI 1.18, 4.38) and poor CSS ( P =0.001, 95% CI 2.45, 27.75). Upon multivariable analysis, a high MRE11 expression was associated with poor CSS ( P =0.019, 95% CI 1.28, 15.65). In summary, MRE11 expression could serve as a potential predictor of prognosis in patients with UTUC.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Urologic Neoplasms , Humans , Carcinoma, Transitional Cell/pathology , Retrospective Studies , Nephroureterectomy/methods , Urologic Neoplasms/diagnosis , Urologic Neoplasms/pathology
16.
J Biol Chem ; 286(33): 28662-28670, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21715338

ABSTRACT

Epidemiologic and clinical research indicates that chronic inflammation increases the risk of certain cancers, possibly through chromosomal instability. However, the mechanism of inflammation-dependent chromosomal instability associated with tumorigenesis is not well characterized. The transcription factor CCAAT/enhancer-binding protein δ (C/EBPδ, CEBPD) is induced by tumor necrosis factor α (TNFα) and expressed in chronically inflamed tissue. In this study, we show that TNFα promotes aneuploidy. Loss of CEBPD attenuated TNFα-induced aneuploidy, and CEBPD caused centromere abnormality. Additionally, TNFα-induced CEBPD expression augmented anchorage-independent growth. We found that TNFα induced expression of aurora kinase C (AURKC) through CEBPD, and that AURKC also causes aneuploidy. Furthermore, high CEBPD expression correlated with AURKC expression in inflamed cervical tissue specimens. These data provide insight into a novel function for CEBPD in inducing genomic instability through the activation of AURKC expression in response to inflammatory signals.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/metabolism , Cervix Uteri/metabolism , Genomic Instability , Protein Serine-Threonine Kinases/biosynthesis , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Uterine Cervicitis/metabolism , Aneuploidy , Animals , Aurora Kinase C , Aurora Kinases , CCAAT-Enhancer-Binding Protein-delta/genetics , Centromere/genetics , Centromere/metabolism , Centromere/pathology , Cervix Uteri/pathology , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology , Uterine Cervicitis/genetics , Uterine Cervicitis/pathology
17.
BMC Mol Cell Biol ; 23(1): 28, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35836107

ABSTRACT

BACKGROUND: In several human cancers, Krüppel-like factor 5 (KLF5), a zinc finger transcription factor, can contribute to both tumor progression or suppression; however, the precise role of KLF5 in nasopharyngeal carcinoma (NPC) remains poorly understood. In this study, the association between KLF5 and microRNA-145-5p (miR-145-5p) in NPC cells was elucidated. RESULTS: Our results showed that KLF5 expression was up-regulated in NPC group compared to normal group. We found that KLF5 exhibited an oncogenic role in NPC cells. The upregulation of miR-145-5p inhibited the proliferation, migration, and invasion of NPC cells. It was observed that miR-145-5p could down-regulate the mRNA and protein expression of KLF5 in NPC cell lines. Additionally, the activity of focal adhesion kinase (FAK), a migration marker, was regulated by miR-145-5p and KLF5 in NPC cells. CONCLUSIONS: The results of this study indicated that miR-145-5p could repress the proliferation, migration, and invasion of NPC cells via KLF5/FAK regulation, and could be a potential therapeutic target for patients with NPC.


Subject(s)
MicroRNAs , Nasopharyngeal Neoplasms , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Transcription Factors/genetics
18.
Cancers (Basel) ; 14(4)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35205628

ABSTRACT

Little is known regarding the molecular characterization of upper tract urothelial carcinoma (UTUC). Novel therapeutic targets and prognostic predictors are imminent. In the present study, we aim to examine the oncogenic function and molecular mechanism of Derlin-1 in UTUC. Derlin-1 overexpression is significantly associated with poor prognosis in patients with UTUC. In vitro, knockdown or over-expression of Derlin-1 markedly regulated UTUC cell invasion and migration. We further discovered miR-375-3p suppresses cell invasion and migration by inversely regulating Derlin-1 and blocking EMT in UTUC cells. Taking this together, miR-375-3p functions as a tumor suppressive microRNA by directly targeting Derlin-1 and blocking epithelial-mesenchymal transition (EMT) in UTUC.

19.
Appl Immunohistochem Mol Morphol ; 30(4): 304-310, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35384881

ABSTRACT

BACKGROUND: Ubiquitin-mediated protein degradation has been reported to be involved in regulating the activity of oncoproteins and tumor suppressors. Dysfunction or dysregulation of the ubiquitin-proteasome system may induce tumorigenesis. Deubiquitinase ubiquitin-specific protease 2a (USP2a) has been reported to regulate cell growth or death and is involved in the pathogenesis of various diseases, including cancers. However, the role of USP2a in upper tract urothelial carcinoma (UTUC) has not been investigated yet. The goal of this study was to evaluate the clinical significance of USP2a expression in UTUC. MATERIALS AND METHODS: A total of 110 UTUC cases were included in this study. USP2a expression level was evaluated through immunohistochemistry staining, and the correlation of USP2a expression level with both clinical and pathologic variables was analyzed. RESULTS: High USP2a expression level was observed in 48 (43.6%) cancer specimens. USP2a expression level was significantly correlated with tumor stage (P=0.001), grade (P=0.033), and tumor recurrence (P=0.008). High USP2a expression level was correlated with poor disease-free survival (P=0.005) and cancer-specific survival (P<0.001). In addition, high USP2a expression level was an independent predictor of poor disease-free survival (hazard ratio=2.31; P=0.007) and cancer-specific survival (hazard ratio=5.49; P=0.009). CONCLUSIONS: This study indicated that USP2a protein expression level may be a potential biomarker for predicting UTUC patient survival. Further prospective studies are needed to investigate the role of USP2a in UTUC progression.


Subject(s)
Carcinoma, Transitional Cell , Ubiquitin Thiolesterase , Urinary Bladder Neoplasms , Humans , Neoplasm Recurrence, Local , Ubiquitin , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases , Urinary Bladder Neoplasms/metabolism
20.
Bioorg Med Chem ; 19(18): 5670-8, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21852140

ABSTRACT

Thirteen anthraquinone derivatives 5-17 including two 3-(3-alkylaminopropoxy)-9,10-anthraquinone (NHA) derivatives 5 and 6, and 11 1-hydroxy-3-(3-alkylaminopropoxy)-9,10-anthraquinone (MHA) derivatives 7-17 were synthesized, evaluated for cytotoxicities against two cancer cell lines, and assayed the generation of reactive oxygen species (ROS) in NTUB1 cells (a human bladder carcinoma cell line). Compound 9 bearing a pyrrolidinyl group induced the stronger cytotoxic effect than those of other synthesized NHA and MHA derivatives. Exposure of NTUB1 cells to 9, 13, and 17 for 24h significantly increased the production of ROS, respectively. Flow cytometric analysis exhibited that the exposure of NTUB1 cells to the selective 9 led to the G2/M phase arrest accompanied by an increase of apoptotic cell death after the incubation for 24h. Compound 9 induced up-regulation of cyclinB1 and p21 expressions. Biological results suggested that the induction of G2/M arrest, apoptosis, and cell death by 9 may associate with increased expression of p21 and cyclin B1, elevation of Bax and p53 levels, and generation of ROS in the cell. In conclusion, these series of compounds may be used as anticancer agents.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , G2 Phase/drug effects , Anthraquinones/chemical synthesis , Anthraquinones/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL