Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Theranostics ; 10(2): 707-724, 2020.
Article in English | MEDLINE | ID: mdl-31903146

ABSTRACT

Muscle-invasive urinary bladder urothelial carcinoma (UBUC) is a lethal disease for which effective prognostic markers and potential therapy targets are still lacking. Previous array comparative genomic hybridization identified that 3q27 is frequently amplified in muscle-invasive UBUCs, one candidate proto-oncogene, B-cell CLL/lymphoma 6 (BCL6), mapped to this region. We therefore aimed to explore its downstream targets and physiological roles in UBUC progression. Methods: Specimens from UBUC patients, NOD/SCID mice and several UBUC-derived cell lines were used to perform quantitative RT-PCR, fluorescence in situ hybridization immunohistochemistry, xenograft, gene stable overexpression/knockdown and a series of in vitro experiments. Results: Amplification of the BCL6 gene lead to upregulation of BCL6 mRNA and protein levels in a substantial set of advanced UBUCs. High BCL6 protein level significantly predicted poor disease-specific and metastasis-free survivals. Knockdown of the BCL6 gene in J82 cells inhibited tumor growth and enhanced apoptosis in the NOD/SCID xenograft model. In vitro experiments demonstrated that BCL6 inhibited cytostasis, induced cell migration, invasion along with alteration of the expression levels of several related regulators. At molecular level, BCL6 inhibited forkhead box O3 (FOXO3) transcription, subsequent translation and upregulation of phosphorylated/inactive FOXO3 through phosphoinositide 3-kinase (PI3K)/AKT serine/threonine kinase (AKT) and/or epidermal growth factor receptor (EGFR)/mitogen-activated protein kinase 1/2 (MAP2K1/2) signaling pathway(s). Two BCL6 binding sites on the proximal promoter region of the FOXO3 gene were confirmed. Conclusion: Overexpression of BCL6 served a poor prognostic factor in UBUC patients. In vivo and in vitro studies suggested that BCL6 functions as an oncogene through direct transrepression of the FOXO3 gene, downregulation and phosphorylation of the FOXO3 protein.


Subject(s)
Adenocarcinoma/pathology , Forkhead Box Protein O3/metabolism , Gene Amplification , Gene Expression Regulation, Neoplastic , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-6/genetics , Urinary Bladder Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Female , Forkhead Box Protein O3/genetics , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Prognosis , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-6/metabolism , Survival Rate , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
Sci Rep ; 9(1): 15745, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31673064

ABSTRACT

The objective of this study was to establish a versatile cell line for replication-incompetent virus production and inactivation with formaldehyde to generate a model of cell-based vaccine manufacturing process. To achieve this goal, we took advantage of the easily accessed chick embryonic fibroblasts. Nine-day old chick embryonic fibroblasts were obtained and subjected to be transduced with a set of lentivirus to develop a chick induced pluripotent stem (ciPS) cell line. Morphological features, positive periodic acid-Schiff staining as well as strong immunocytofluorescence of alkaline phosphatase, intestinal (ALPI) and POU class 5 homeobox 1 (POU5F1) proteins suggested that these chick embryonic fibroblasts have been transformed into ciPS cells. Further differentiation and immunocytofluorescence assays confirmed that this ciPS cell line possesses capacities and potentials to form embryoid bodies, differentiate into all three embryonic layers: ectoderm, mesoderm and endoderm with evidence of strongly positive and specific molecular markers. Immunoblot analysis next demonstrated that through recombinant DNA technology and the 2nd generation lentiviral transfer system, the goose hemagglutinin gene (H5) gene was packaged into the replication-incompetent virus and highly expressed in a bladder cancer-derived cell line, T24, after transduction. The titer of ciPS-generated replication-incompetent virus is comparable to that from the Phoenix-AMPHO cell line, which is a commercial and high productive retrovirus producer. Our study successfully established a ciPS cell line which is able to produce replication-incompetent virus, providing a new strategy for cell-based vaccine production after virus inactivation.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Lentivirus/genetics , Plasmids/metabolism , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Chick Embryo , Chickens , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Geese , Hemagglutinins/genetics , Hemagglutinins/metabolism , Induced Pluripotent Stem Cells/cytology , Lentivirus/physiology , Octamer Transcription Factor-3/metabolism , Plasmids/genetics , Taiwan , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Vaccines, Synthetic/metabolism , Virus Replication
3.
J Cell Physiol ; 234(6): 9551-9563, 2019 06.
Article in English | MEDLINE | ID: mdl-30367486

ABSTRACT

The objective was to investigate the upstream mechanisms of apoptosis which were triggered by a novel antimicrotubule drug, ABT-751, in a tumor protein p53 ( TP53)-deficient hepatocellular carcinoma-derived Hep-3B cells. A series of in vitro assays indicated that ABT-751 caused the disruption of the mitotic spindle structure, collapse of mitochondrial membrane potential, generation of reactive oxygen species, DNA damage, G 2 /M cell cycle arrest, inhibition of anchorage-independent cell growth and apoptosis in Hep-3B cells accompanied by alteration of the expression levels of several DNA damage checkpoint proteins and cell cycle regulators. Subsequently, ABT-751 triggered apoptosis along with markedly upregulated several proapoptotic proteins involving in extrinsic, intrinsic, and caspase-mediated apoptotic pathways. A pan-caspase inhibitor suppressed ABT-751-induced apoptosis. ABT-751 also induced autophagy soon after the occurrence of apoptosis through the suppression of AKT serine/threonine kinase/mechanistic target of rapamycin signaling pathway. Exogenous expression of the TP53 gene significantly incurred both apoptosis and autophagy in Hep-3B cells. Pharmacological inhibition of autophagosome (early autophagy) but not autolysosome (late autophagy) enhanced ABT-751-induced apoptosis in TP53-deficient Hep-3B cells. Our study provided a new strategy to augment ABT-751-induced apoptosis in TP53-deficient cells.


Subject(s)
Apoptosis/drug effects , Autophagosomes/metabolism , Lysosomes/metabolism , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/deficiency , Autophagosomes/drug effects , Autophagy/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Humans , Lysosomes/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitosis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sulfonamides/chemistry , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics
4.
Clin Cancer Res ; 23(24): 7650-7663, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28972042

ABSTRACT

Purpose: Urinary bladder urothelial carcinoma (UBUC) is a common malignant disease in developed countries. Cell-cycle dysregulation resulting in uncontrolled cell proliferation has been associated with UBUC development. This study aimed to explore the roles of TMCO1 in UBUCs.Experimental Design: Data mining, branched DNA assay, immunohistochemistry, xenograft, cell culture, quantitative RT-PCR, immunoblotting, stable and transient transfection, lentivirus production and stable knockdown, cell-cycle, cell viability and proliferation, soft-agar, wound-healing, transwell migration and invasion, coimmunoprecipitation, immunocytochemistry, and AKT serine/threonine kinase (AKT) activity assays and site-directed mutagenesis were used to study TMCO1 involvement in vivo and in vitroResults: Data mining identified that the TMCO1 transcript was downregulated during the progression of UBUCs. In distinct UBUC-derived cell lines, changes in TMCO1 levels altered the cell-cycle distribution, cell viability, cell proliferation, and colony formation and modulated the AKT pathway. TMCO1 recruited the PH domain and leucine-rich repeat protein phosphatase 2 (PHLPP2) to dephosphorylate pAKT1(serine 473) (S473). Mutagenesis at S60 of the TMCO1 protein released TMCO1-induced cell-cycle arrest and restored the AKT pathway in BFTC905 cells. Stable TMCO1 (wild-type) overexpression suppressed, whereas T33A and S60A mutants recovered, tumor size in xenograft mice.Conclusions: Clinical associations, xenograft mice, and in vitro indications provide solid evidence that the TMCO1 gene is a novel tumor suppressor in UBUCs. TMCO1 dysregulates cell-cycle progression via suppression of the AKT pathway, and S60 of the TMCO1 protein is crucial for its tumor-suppressor roles. Clin Cancer Res; 23(24); 7650-63. ©2017 AACR.


Subject(s)
Carcinoma/genetics , Membrane Proteins/genetics , Oncogene Protein v-akt/genetics , Phosphoprotein Phosphatases/genetics , Tumor Suppressor Proteins/genetics , Animals , Calcium Channels , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Data Mining , Humans , Lentivirus/genetics , Mice , Mutagenesis , Signal Transduction/genetics , Urinary Bladder/pathology , Urothelium/pathology , Xenograft Model Antitumor Assays
5.
Toxicol Appl Pharmacol ; 311: 88-98, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27678524

ABSTRACT

The objective was to investigate the upstream mechanisms of apoptosis which were triggered by a novel anti-microtubule drug, ABT-751, in hepatocellular carcinoma-derived Huh-7 cells. Effects of ABT-751 were evaluated by immunocytochemistry, flow cytometric, alkaline comet, soft agar, immunoblotting, CytoID, green fluorescent protein-microtubule associated protein 1 light chain 3 beta detection, plasmid transfection, nuclear/cytosol fractionation, coimmunoprecipitation, quantitative reverse transcription-polymerase chain reaction, small-hairpin RNA interference and mitochondria/cytosol fractionation assays. Results showed that ABT-751 caused dysregulation of microtubule, collapse of mitochondrial membrane potential, generation of reactive oxygen species (ROS), DNA damage, G2/M cell cycle arrest, inhibition of anchorage-independent cell growth and apoptosis in Huh-7 cells. ABT-751 also induced early autophagy via upregulation of nuclear TP53 and downregulation of the AKT serine/threonine kinase (AKT)/mechanistic target of rapamycin (MTOR) pathway. Through modulation of the expression levels of DNA damage checkpoint proteins and G2/M cell cycle regulators, ABT-751 induced G2/M cell cycle arrest. Subsequently, ABT-751 triggered apoptosis with marked downregulation of B-cell CLL/lymphoma 2, upregulation of mitochondrial BCL2 antagonist/killer 1 and BCL2 like 11 protein levels, and cleavages of caspase 8 (CASP8), CASP9, CASP3 and DNA fragmentation factor subunit alpha proteins. Suppression of ROS significantly decreased ABT-751-induced autophagic and apoptotic cells. Pharmacological inhibition of autophagy significantly increased the percentages of ABT-751-induced apoptotic cells. The autophagy induced by ABT-751 plays a protective role to postpone apoptosis by exerting adaptive responses following microtubule damage, ROS and/or impaired mitochondria.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Microtubules/drug effects , Sulfonamides/pharmacology , Caspases/metabolism , Cell Division/drug effects , Cell Line, Tumor , G2 Phase/drug effects , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Oncotarget ; 6(11): 9220-39, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25940704

ABSTRACT

In this study, we report that EMP2 plays a tumor suppressor role by inducing G2/M cell cycle arrest, suppressing cell viability, proliferation, colony formation/anchorage-independent cell growth via regulation of G2/M checkpoints in distinct urinary bladder urothelial carcinoma (UBUC)-derived cell lines. Genistein treatment or exogenous expression of the cAMP responsive element binding protein 1 (CREB1) gene in different UBUC-derived cell lines induced EMP2 transcription and subsequent translation. Mutagenesis on either or both cAMP-responsive element(s) dramatically decreased the EMP2 promoter activity with, without genistein treatment or exogenous CREB1 expression, respectively. Significantly correlation between the EMP2 immunointensity and primary tumor, nodal status, histological grade, vascular invasion and mitotic activity was identified. Multivariate analysis further demonstrated that low EMP2 immunoexpression is an independent prognostic factor for poor disease-specific survival. Genistein treatments, knockdown of EMP2 gene and double knockdown of CREB1 and EMP2 genes significantly inhibited tumor growth and notably downregulated CREB1 and EMP2 protein levels in the mice xenograft models. Therefore, genistein induced CREB1 transcription, translation and upregulated pCREB1(S133) protein level. Afterward, pCREB1(S133) transactivated the tumor suppressor gene, EMP2, in vitro and in vivo. Our study identified a novel transcriptional target, which plays a tumor suppressor role, of CREB1.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Genistein/pharmacology , Membrane Glycoproteins/genetics , Transcriptional Activation/genetics , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival , Cyclic AMP Response Element-Binding Protein/biosynthesis , Female , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering , Transplantation, Heterologous , Urinary Bladder/pathology , Urinary Bladder Neoplasms/mortality
7.
Toxicol Rep ; 2: 322-332, 2015.
Article in English | MEDLINE | ID: mdl-28962365

ABSTRACT

Cell cycle deregulation is common in human hepatocellular carcinoma (HCC). To ensure proper cell cycle controlling, cyclin/cyclin-dependent kinases (CDK) complexes are tightly regulated by CDK inhibitors (CKIs) in normal cells. However, insufficient cyclin-dependent kinase inhibitor 1B (CDKN1B, also known as p27Kip1) and CDKN1C (p57Kip2) proteins are characteristics of high-risk HCC. In two HCC-derived cell lines with distinct genetic backgrounds, we identified a small natural compound, goniothalamin (GTN), serving as an inducer of CKIs. In TP53-mutated (Y220C) and retinoblastoma 1 (RB1)-positive Huh-7 cells, GTN stabilized CDKN1B protein levels by targeting the degradation of its specific E3 ubiquitin ligase (S-phase kinase-associated protein 2). Alternatively, in TP53- and RB1-negative Hep-3B cells, GTN increased CDKN1C transcription and its subsequent translation by acting as a histone deacetylase inhibitor. In both cell lines, GTN induced G0/G1 cell cycle arrest, delayed S phase entry of cells and inhibited anchorage-independent cell growth which might be attributed to the upregulation of CKIs and downregulation of several positive cell cycle regulators, including CDC28 protein kinase regulator subunit 1B, cyclin E1 and D1, cyclin-dependent kinase 2 (CDK2), CDK4, CDK6, E2F transcription factor 1 and/or transcription factor Dp-1. Therefore, GTN might represent a novel class of anticancer drug that induces CKIs through post-translational and epigenetic modifications.

8.
Ann Surg Oncol ; 21 Suppl 4: S711-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24889487

ABSTRACT

BACKGROUND: Attenuated endogenous protein levels of cyclin-dependent kinase 2 associated protein 1 (p12(CDK2AP1)) and its active homodimer p25(CDK2AP1) were found in myxofibrosarcoma-derived cell lines. Clinical and biological significances of this putative tumor suppressor in myxofibrosarcoma were studied. METHODS: Plasmids carrying the CDK2AP1 gene and small hairpin RNA interference (shRNAi) targeting CDK2AP1 were transfected into NMFH-1 and/or OH931 cells to evaluate the effects on the CDK2, active caspase 3 (CASP3), cleaved-CASP8 and -CASP9 levels, cell cycle regulation, and/or apoptotic responses. Immunostaining of p12(CDK2AP1) was interpretable in 102 primary myxofibrosarcomas and correlated with clinicopathological variables, CDK2, Ki-67 and active CASP3 protein levels, and disease-specific survival. RESULTS: Exogenous expression of p12(CDK2AP1) in NMFH-1 and OH931 cells significantly induced G0/G1 cell cycle arrest and down-regulated CDK2 protein level. In NMFH-1 cells, these aspects were reversed by shRNAi targeting CDK2AP1 gene. Increased active CASP3 and cleaved-CASP9, but not -CASP8, were detected after CDK2AP1 overexpression, suggesting the cellular apoptosis were induced through the mitochondrial pathway. Immunostains of p12(CDK2AP1) were aberrantly decreased in 56.9 % of cases; positively and negatively correlated with protein levels of CDK2 (p = 0.023), Ki-67 (p = 0.001) and active CASP3 (p < 0.001), respectively. Following by high histological grades, p12(CDK2AP1) down-regulation was predictive of worse disease-specific survival in univariate (p = 0.003) and multivariate (p = 0.004) analyses. CONCLUSIONS: Through down-regulation of CDK2, high p12(CDK2AP1) level induced cell cycle arrest and the mitochondrial-dependent apoptotic pathway. Low p12(CDK2AP1) level represents a poor prognostic factor in patients with myxofibrosarcoma.


Subject(s)
Fibrosarcoma/enzymology , Fibrosarcoma/genetics , Mitochondria/metabolism , Myxosarcoma/enzymology , Myxosarcoma/genetics , Tumor Suppressor Proteins/genetics , Apoptosis/genetics , Caspase 3/analysis , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Survival , Chromobox Protein Homolog 5 , Cyclin-Dependent Kinase 2/analysis , Cyclin-Dependent Kinase 2/metabolism , Female , Fibrosarcoma/chemistry , Humans , Ki-67 Antigen/analysis , Male , Middle Aged , Mitotic Index , Myxosarcoma/chemistry , Plasmids , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Survival Rate , Transfection
9.
Tumour Biol ; 35(3): 2619-29, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24218338

ABSTRACT

Data mining on public domain identified that stathmin 1 (STMN1) transcript was significantly higher expressed in nasopharyngeal carcinoma (NPC). Also known as the oncoprotein 18, STMN1 performs an important function in regulating rapid microtubule remodeling of the cytoskeleton in response to the cellular conditions. Immunoexpression of STMN1 was retrospectively assessed in biopsies of 124 consecutive NPC patients without initial distant metastasis and treated with consistent guidelines. The outcome was correlated with clinicopathological features and patient survivals. Results indicated that high STMN1 expressions (50 %) were correlated with advanced age (p = 0.027), higher T stage (p = 0.003), and overall clinical stage (p = 0.006) by the 7th American Joint Committee of Cancer Staging. In multivariate analyses, high STMN1 expression emerged as an independent prognosticator for worse disease-specific survival (p = 0.001), distal metastasis-free survival (p = 0.003), and local recurrence-free survival (p = 0.006). Exogenous expression of E2F transcription factor 1 (E2F1) or/and its dimeric partner, transcription factor Dp-1 (TFDP1), notably induced the STMN1 protein level in a NPC-derived cell line, TW01. Accordingly, high STMN1 protein level is commonly associated with adverse prognosticators and confers tumor aggressiveness in patients with NPC, and its upregulation might be attributed to E2F1 and/or TFDP1 transactivation.


Subject(s)
Biomarkers, Tumor/analysis , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/mortality , Stathmin/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma , Disease-Free Survival , Female , Humans , Immunoblotting , Immunohistochemistry , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Neoplasm Staging , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Stathmin/analysis , Transfection , Young Adult
10.
Clin Cancer Res ; 19(11): 2861-72, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23549872

ABSTRACT

PURPOSE: The principal goals were to identify and validate targetable metabolic drivers relevant to myxofibrosarcoma pathogenesis using a published transcriptome. EXPERIMENTAL DESIGN: As the most significantly downregulated gene regulating amino acid metabolism, argininosuccinate synthetase (ASS1) was selected for further analysis by methylation-specific PCR, pyrosequencing, and immunohistochemistry of myxofibrosarcoma samples. The roles of ASS1 in tumorigenesis and the therapeutic relevance of the arginine-depriving agent pegylated arginine deiminase (ADI-PEG20) were elucidated in ASS1-deficient myxofibrosarcoma cell lines and xenografts with and without stable ASS1 reexpression. RESULTS: ASS1 promoter hypermethylation was detected in myxofibrosarcoma samples and cell lines and was strongly linked to ASS1 protein deficiency. The latter correlated with increased tumor grade and stage and independently predicted a worse survival. ASS1-deficient cell lines were auxotrophic for arginine and susceptible to ADI-PEG20 treatment, with dose-dependent reductions in cell viability and tumor growth attributable to cell-cycle arrest in the S-phase. ASS1 expression was restored in 2 of 3 ASS1-deficient myxofibrosarcoma cell lines by 5-aza-2'-deoxycytidine, abrogating the inhibitory effect of ADI-PEG20. Conditioned media following ASS1 reexpression attenuated HUVEC tube-forming capability, which was associated with suppression of MMP-9 and an antiangiogenic effect in corresponding myxofibrosarcoma xenografts. In addition to delayed wound closure and fewer invading cells in a Matrigel assay, ASS1 reexpression reduced tumor cell proliferation, induced G1-phase arrest, and downregulated cyclin E with corresponding growth inhibition in soft agar and xenograft assays. CONCLUSIONS: Our findings highlight ASS1 as a novel tumor suppressor in myxofibrosarcomas, with loss of expression linked to promoter methylation, clinical aggressiveness, and sensitivity to ADI-PEG20.


Subject(s)
Argininosuccinate Synthase/genetics , Fibrosarcoma/genetics , Genes, Tumor Suppressor , Leiomyosarcoma/genetics , Adult , Aged , Amino-Acid N-Acetyltransferase/biosynthesis , Animals , Argininosuccinate Synthase/deficiency , Argininosuccinate Synthase/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/drug effects , Cluster Analysis , DNA Methylation , Female , Fibrosarcoma/drug therapy , Fibrosarcoma/mortality , Fibrosarcoma/pathology , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Hydrolases/administration & dosage , Hydrolases/pharmacology , Leiomyosarcoma/drug therapy , Leiomyosarcoma/mortality , Leiomyosarcoma/pathology , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Neovascularization, Pathologic/genetics , Phenotype , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacology , Prognosis , Xenograft Model Antitumor Assays
11.
J Clin Pathol ; 66(7): 573-82, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23539739

ABSTRACT

BACKGROUND: Urothelial carcinoma (UC) is prevalent worldwide. Dysregulation of cell growth is a critical event of tumorigenesis and has not been assessed systemically in UC. We thus assessed the published transcriptome of urinary bladder urothelial carcinoma (UBUC) and identified insulin-like growth factor-binding protein-5 (IGFBP-5) as the most significantly upregulated gene associated with the regulation of cell growth. Moreover, validated by using public domain data set, IGFBP-5 expression also significantly predicted worse outcome. IGFBP-5 is one of the binding proteins that regulate insulin-like growth factors (IGFs) and its significance has not been comprehensively evaluated in UCs. METHODS: Using immunohistochemistry, we evaluated the IGFBP-5 expression status and its associations with clinicopathological features and survival in 340 cases of upper urinary tract urothelial carcinoma (UTUC) and 295 cases of UBUC. Western blot analysis was used to evaluate IGFBP-5 protein expression in human urothelial cell (HUC) lines. RESULTS: IGFBP-5 overexpression was significantly associated with advanced pT stage (p<0.001), high histological grade (UTUC, p<0.001; UBUC, p=0.035), lymph node metastasis (UTUC, p=0.006; UBUC, p=0.004), vascular invasion (UTUC, p<0.001; UBUC, p=0.003), perineural invasion (UTUC, p=0.034; UBUC, p=0.021) and frequent mitosis (UTUC, p<0.001; UBUC, p=0.023). IGFBP-5 overexpression also independently predicted poor disease-specific survival and metastasis-free survival in both groups of patients. Western blot analysis showed IGFBP-5 protein as overexpressed in human urothelial cancer cell lines and not in normal urothelial cancer cells. CONCLUSIONS: IGFBP-5 plays an important role in tumour progression in UC. Its overexpression is associated with advanced tumour stage and conferred poorer clinical outcome.


Subject(s)
Carcinoma, Transitional Cell/secondary , Insulin-Like Growth Factor Binding Protein 5/metabolism , Kidney Neoplasms/pathology , Urinary Bladder Neoplasms/pathology , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/mortality , Cell Line, Tumor , Disease Progression , Female , Gene Expression , Gene Expression Profiling , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Male , Middle Aged , Prognosis , Survival Rate , Taiwan/epidemiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality
12.
Ann Surg Oncol ; 20(12): 4041-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22911364

ABSTRACT

BACKGROUND: Through data mining the Stanford Microarray Database, the stathmin 1 (STMN1) transcript was found to be frequently upregulated in the hepatocellular carcinoma (HCC) with low alpha-fetoprotein level. The molecular mechanism of STMN1 upregulation in HCCs remained unclear. METHODS: Quantitative RT-PCR, immunoblotting, immunohistochemistry, and transfection of expression or small hairpin RNA interference plasmids, chromatin immunoprecipitation (ChIP), and quantitative ChIP assays were performed in HCC specimens or 2 distinct HCC-derived cell lines. Dual luciferase assay and site-directed mutagenesis were applied to analyze the activities of STMN1 proximal promoter region. RESULTS: STMN1 mRNA and proteins were significantly associated with several clinicopathological features. High STMN1 or E2F1 immunoexpression was predictive of poor overall survival (OS) rate (P < .01). In HCC-derived cell lines, E2F1 was elevated before STMN1 mRNA during the cell cycle. Exogenous expression of E2F1 or both transcription factor DP-1 (TFDP1) and E2F1 genes induced E2F1 and STMN1 mRNA (P < .01). Knockdown of the E2F1 gene suppressed E2F1 and STMN1 mRNA and E2F1 and STMN1 protein levels (P < .05). The promoter activity of STMN1 gene increased with overexpression of both E2F1 and TFDP1 genes (P < .05); however, it decreased when mutations were introduced in the E2F1-binding sites (P < .05). CONCLUSIONS: Upregulation of E2F1 and STMN1 proteins associate with worse outcomes in patients with HCC. E2F1 significantly correlates with STMN1 protein level in HCC lesions and in vitro transactivation assays, suggesting that STMN1 gene is transactivated by the E2F1 protein.


Subject(s)
Carcinoma, Hepatocellular/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver/metabolism , Stathmin/genetics , Transcriptional Activation , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Proliferation , Chromatin Immunoprecipitation , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/genetics , Female , Flow Cytometry , Follow-Up Studies , Humans , Immunoenzyme Techniques , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Luciferases/metabolism , Male , Mutagenesis, Site-Directed , Mutation/genetics , Neoplasm Staging , Prognosis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Stathmin/metabolism , Transcription Factor DP1/genetics , Transcription Factor DP1/metabolism , Tumor Cells, Cultured , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
13.
BMC Cancer ; 12: 611, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23259573

ABSTRACT

BACKGROUND: HuR is an RNA-binding protein that post-transcriptionally modulates the expressions of various target genes implicated in carcinogenesis, such as CCNA2 encoding cyclin A. No prior study attempted to evaluate the significance of HuR expression in a large cohort with upper urinary tract urothelial carcinomas (UTUCs). METHODS: In total, 340 cases of primary localized UTUC without previous or concordant bladder carcinoma were selected. All of these patients received ureterectomy or radical nephroureterectomy with curative intents. Pathological slides were reviewed, and clinical findings were collected. Immunostaining for HuR and cyclin A was performed and evaluated by using H-score. The results of cytoplasmic HuR and nuclear cyclin A expressions were correlated with disease-specific survival (DSS), metastasis-free survival (MeFS), urinary bladder recurrence-free survival (UBRFS), and various clinicopathological factors. RESULTS: HuR cytoplasmic expression was significantly related to the pT status, lymph node metastasis, a higher histological grade, the pattern of invasion, vascular and perineurial invasion, and cyclin A expression (p = 0.005). Importantly, HuR cytoplasmic expression was strongly associated with a worse DSS (p < 0.0001), MeFS (p < 0.0001), and UBRFS (p = 0.0370) in the univariate analysis, and the first two results remained independently predictive of adverse outcomes (p = 0.038, relative risk [RR] = 1.996 for DSS; p = 0.027, RR = 1.880 for MeFS). Cyclin A nuclear expression was associated with a poor DSS (p = 0.0035) and MeFS (p = 0.0015) in the univariate analysis but was not prognosticatory in the multivariate analyses. High-risk patients (pT3 or pT4 with/without nodal metastasis) with high HuR cytoplasmic expression had better DSS if adjuvant chemotherapy was performed (p = 0.015). CONCLUSIONS: HuR cytoplasmic expression was correlated with adverse phenotypes and cyclin A overexpression and also independently predictive of worse DSS and MeFS, suggesting its roles in tumorigenesis or carcinogenesis and potentiality as a prognostic marker of UTUC. High HuR cytoplasmic expression might identify patients more likely to be beneficial for adjuvant chemotherapy.


Subject(s)
Carcinoma, Transitional Cell/metabolism , Cyclin A/metabolism , Cytoplasm/metabolism , ELAV Proteins/metabolism , Urologic Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/therapy , Cohort Studies , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Urologic Neoplasms/pathology , Urologic Neoplasms/therapy
14.
J Clin Pathol ; 65(9): 795-801, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22791769

ABSTRACT

BACKGROUND AND AIM: Low expression of cyclin-dependent kinase 2-associated protein (CDK2AP1) is associated with tumour progression in oral and oesophageal carcinomas, but is not well studied in patients with head and neck cancer and nasopharyngeal carcinoma (NPC). METHODS: A rabbit anti-human CDK2AP1 polyclonal antibody was prepared. Immunoblotting of CDK2AP1 was examined in three cell lines and immunoexpression was retrospectively assessed in biopsies of 124 consecutive NPC patients without initial distant metastasis and treated with consistent guidelines. RESULTS: Higher CDK2AP1 expression level was identified in dysplastic oral keratinocytes, compared with two NPC-derived HONE-1 and TW01 cell lines. Low expression of CDK2AP1 (50.8%) was correlated with advanced nodal status (p=0.002) and American Joint Committee on Cancer (AJCC) stage (p=0.004). In multivariate analyses, low CDK2AP1 expression emerged as an independent prognosticator for worse disease-specific survival (DSS; p=0.037) and local recurrence-free survival (LRFS; p=0.042), along with AJCC stage III-IV (p=0.034, DSS; p=0.029, LRFS). CONCLUSIONS: Low CDK2AP1 expression is common and associated with adverse prognosticators, conferring tumour aggressiveness through cycle cycle, cell growth or apoptosis cellular processes.


Subject(s)
Biomarkers, Tumor/metabolism , Nasopharyngeal Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Biomarkers, Tumor/genetics , Biopsy , Blotting, Western , Carcinoma , Cell Line, Tumor , Chi-Square Distribution , Disease Progression , Disease-Free Survival , Down-Regulation , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/therapy , Neoplasm Recurrence, Local , Neoplasm Staging , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Assessment , Risk Factors , Transfection , Tumor Suppressor Proteins/genetics
15.
J Clin Pathol ; 65(9): 802-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22685262

ABSTRACT

BACKGROUNDS: Urothelial carcinoma of the urinary bladder (UCUB) is prevalent in developed countries. It often shows genetic instability and is associated with amplification (or gain) of various oncogenic genes or suppressive genes. Rsf-1, a subunit of ATP-dependent chromatin-remodelling complexes that mediates ATPase-dependent chromatin remodelling, confers tumour aggressiveness in certain carcinomas. The authors evaluate the Rsf-1 gene and expression status and its associations with clinicopathological features and survival in their UCUB collection. METHODS: Immunohistochemistry was used to assess the Rsf-1 expression profile in 295 UCUB specimens, and was found to correlate with clinicopathological data. Real-time RT-PCR and fluorescence in situ hybridisation were used to detect RSF-1 mRNA expression and gene dosage in 20 independent cases. Western blot analysis was used to evaluate Rsf-1 protein expression in human urothelial cell lines. RESULTS: Rsf-1 overexpression was demonstrated in 101 cases (34.2%), and was significantly associated with advanced primary tumour (p<0.001), nodal metastasis (p=0.004), higher histological grades (p=0.001) and frequent mitoses (p<0.001). Moreover, it was predictive in disease-specific survival and metastasis-free survival in both univariate and multivariate analyses (p<0.0001 for both). Although RSF-1 gene amplification can be barely detected, its mRNA expression is significantly enhanced in tumours with higher primary tumour (p=0.041) and positive nodal statuses (p=0.010), respectively. Rsf-1 protein was abundant in invasive urothelial carcinoma cells but was not benign. CONCLUSIONS: Overexpression of Rsf-1 is associated with higher tumour stage and poorer clinical outcome. The current study by the authors suggests gene amplification-independent mechanisms driving Rsf-1 overexpression during UCUB tumour progression.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Gene Amplification , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Urinary Bladder Neoplasms/metabolism , Urothelium/metabolism , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma/genetics , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/therapy , Cell Line, Tumor , Chi-Square Distribution , Disease-Free Survival , Female , Gene Dosage , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Nuclear Proteins/genetics , Prognosis , Proportional Hazards Models , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Retrospective Studies , Risk Assessment , Risk Factors , Trans-Activators/genetics , Up-Regulation , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Urothelium/pathology
16.
J Clin Pathol ; 65(8): 687-92, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22569540

ABSTRACT

AIMS: Neoadjuvant chemoradiation therapy (CRT) is an increasingly used therapeutic strategy for rectal cancer. Clinically, it remains a major challenge to predict therapeutic response and patient outcome after CRT. Rsf-1 (HBXAP), a novel nuclear protein with histone chaperon function, mediates ATPase-dependent chromatin remodelling and confers tumour aggressiveness and predicts therapeutic response in certain carcinomas. However, the expression of Rsf-1 has never been reported in rectal cancer. This study examined the predictive and prognostic impacts of Rsf-1 expression in patients with rectal cancer following neoadjuvant CRT. METHODS: Rsf-1 immunoexpression was retrospectively assessed for pre-treatment biopsies of 172 rectal cancer patients without initial distant metastasis. All of them were treated with neoadjuvant CRT followed by surgery. The results were correlated with the clinicopathological features, therapeutic response, tumour regression grade and metastasis-free survival (MeFS), local recurrent-free survival and disease-specific survival. RESULTS: Present in 82 cases (47.7%), high-expression of Rsf-1 was associated with advanced pre-treatment tumour status (T3, T4, p=0.020), advanced post-treatment tumour status (T3, T4, p<0.001) and inferior tumour regression grade (p=0.028). Of note, high-expression of Rsf-1 emerged as an adverse prognosticator for diseases-specific survival (p=0.0092) and significantly predicted worse MeFS (p=0.0006). Moreover, high-expression of Rsf-1 also remained prognostic independent for worse MeFS (HR 2.834; p=0.0214). CONCLUSIONS: High-expression of Rsf-1 is associated with poor therapeutic response and adverse outcome in rectal cancer patients treated with neoadjuvant CRT, which confers tumour aggressiveness and therapeutic resistance through chromatin remodelling and represents a potential prognostic biomarker in rectal cancer.


Subject(s)
Adenocarcinoma/chemistry , Adenocarcinoma/therapy , Biomarkers, Tumor/analysis , Chemoradiotherapy, Adjuvant , Neoadjuvant Therapy , Nuclear Proteins/analysis , Rectal Neoplasms/chemistry , Rectal Neoplasms/therapy , Trans-Activators/analysis , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Aged , Biopsy , Chi-Square Distribution , Digestive System Surgical Procedures , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Staging , Predictive Value of Tests , Rectal Neoplasms/mortality , Rectal Neoplasms/pathology , Retrospective Studies , Risk Assessment , Risk Factors , Taiwan , Time Factors , Treatment Outcome , Up-Regulation
17.
BMJ Open ; 2(2): e000900, 2012.
Article in English | MEDLINE | ID: mdl-22492389

ABSTRACT

OBJECTIVE: To evaluate the expression of epithelial membrane protein-2 (EMP2) protein and its clinicopathological associations in patients with nasopharyngeal carcinoma. DESIGN: Retrospective population-based cohort study. SETTING: This study was based on a biobank in Chi-Mei Medical Center (Tainan, Taiwan) from 1993 to 2002. PARTICIPANTS: Biopsies of 124 consecutive nasopharyngeal carcinoma patients without initial distant metastasis and treated with consistent guidelines were assessed. Immunoexpressions of EMP2 were analysed and the outcomes were correlated with clinicopathological features and patient survivals. PRIMARY AND SECONDARY OUTCOME MEASURES: Immunoexpressions of EMP2 were analyzed and the outcomes were correlated with clinicopathological features and patient survivals. RESULTS: Loss of EMP2 expression (49.2%) was correlated with advanced primary tumour (p=0.044), nodal status (p=0.045) and the 7th American Joint Committee on Cancer stage (p=0.027). In multivariate analyses, loss of EMP2 expression emerged as an independent prognosticator for worse disease-specific survival (DSS; p=0.015) and local recurrence-free survival (LRFS; p=0.030), along with the American Joint Committee on Cancer stages III-IV (p=0.034, DSS; p=0.023, LRFS). CONCLUSIONS: Loss of EMP2 expression is common and associated with adverse prognosticators and might confer tumour aggressiveness through hampering its interaction with specific membrane protein(s) and hence the downstream signal transduction pathway(s).

18.
Ann Surg Oncol ; 19(8): 2716-25, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22476749

ABSTRACT

BACKGROUND: Myxofibrosarcoma is genetically complex and remains obscure in molecular determinants of clinical aggressiveness. Our prior study revealed recurrent gains of 7q in myxofibrosarcomas where MET and CDK6 genes displayed increased DNA copies. Previously, we demonstrated the implication of MET overexpression, prompting us to further elucidate the roles of CDK6 in myxofibrosarcomas. MATERIALS: On tissue microarrays, CDK6 immunoexpression was assessable in 77 primary tumors, 55 of which were successfully quantified for CDK6 and MET genes by real-time PCR using genomic DNA extracted from laser-microdissected tumor cells. Gene status and protein expression of CDK6 were correlated with each other, clinicopathological variables, metastasis-free survival (MFS), and disease-specific survival (DSS). RESULTS: Protein overexpression and gene amplification of CDK6, which were detected in 21 of 77 (27.2 %) and 13 of 55 cases (23.6 %), respectively, were highly related to each other (p < .001) and associated with higher grades (overexpression, p = .004; amplification, p = .014). There was a strong correlation between CDK6 and MET gene copies (p < .001, r = 0.0714). Importantly, CDK6 protein overexpression (MFS, p = .0002; DSS, p = .0015) and gene amplification (MFS, p = .0001; DSS, p = .0083) were both univariately associated with worse outcomes. Together with nonextremity location and AJCC stage III disease, CDK6 overexpression independently portended inferior MFS (p = .0015, risk ratio [RR] = 7.411). This aberration, along with nonextremity location, was also an independent adverse prognosticator of DSS (p = .0069, RR = 6.006). CONCLUSIONS: In approximately a quarter of primary myxofibrosarcomas, CDK6 overexpression is mostly driven by gene amplification on 7q, associated with adverse prognosticators, and independently predictive of worse outcomes, highlighting its possible causative role in tumor aggressiveness.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Cyclin-Dependent Kinase 6/genetics , Fibrosarcoma/genetics , Gene Amplification , Myxosarcoma/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase 6/metabolism , DNA, Neoplasm/genetics , Female , Fibrosarcoma/mortality , Fibrosarcoma/pathology , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Middle Aged , Myxosarcoma/mortality , Myxosarcoma/pathology , Neoplasm Grading , Neoplasm Staging , Prognosis , Prospective Studies , Real-Time Polymerase Chain Reaction , Survival Rate , Tissue Array Analysis
19.
Clin Cancer Res ; 18(6): 1598-610, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22322669

ABSTRACT

PURPOSE: Myxofibrosarcoma remains obscure in molecular determinants of clinical aggressiveness, for which we elucidated implications of SKP2 amplification. EXPERIMENTAL DESIGN: Array comparative genomic hybridization was applied on samples and cell lines (NMFH-1 to OH931) to search causal genes of tumor progression. SKP2 gene dosage was determined in 82 independent tumors for clinical correlates. Stable SKP2 knockdown was achieved in myxofibrosarcoma cells to assess its oncogenic attributes and candidate mediators in prometastatic function. Pharmacologic assays were evaluated in vitro and in vivo for the therapeutic relevance of bortezomib. RESULTS: DNA gains frequently involved 5p in which three amplicons were differentially overrepresented in samples behaving unfavorably, encompassing mRNA-upregulated TRIO, SKP2, and AMACR genes. Detected in NMFH-1 cells and 38% of tumors, SKP2 amplification was associated with SKP2 immunoexpression and adverse prognosticators and independently predictive of worse outcomes. Nevertheless, SKP2-expressing OH931 cells and 14% of such tumors lacked gene amplification. Knockdown of SKP2 suppressed proliferation, anchorage-independent growth, migration, and invasion of sarcoma cells and downregulated motility-promoting genes, including ITGB2, ACTN1, IGF1, and ENAH. In vitro, bortezomib downregulated SKP2 expression at the mRNA level with p27(kip1) accumulation, induced caspase activation, and decreased cell viability in myxofibrosarcoma cells but not in fibroblasts. In vivo, bortezomib inhibited growth of NMFH-1 xenografts, the cells of which displayed decreased SKP2 expression but increased p27(kip1) and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). CONCLUSIONS: As a predominant mechanism driving protein overexpression, SKP2 amplification confers tumor aggressiveness in myxofibrosarcoma. The sensitivity of myxofibrosarcoma cells to bortezomib with SKP2-repressing effect indicates the potentiality of ubiquitin-proteasome pathway as a therapeutic target.


Subject(s)
Fibrosarcoma/genetics , Gene Amplification , S-Phase Kinase-Associated Proteins/genetics , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Comparative Genomic Hybridization , Female , Gene Dosage , Guanine Nucleotide Exchange Factors/genetics , Humans , Male , Mice , Mice, SCID , Middle Aged , Prognosis , Protein Serine-Threonine Kinases/genetics , Pyrazines/pharmacology , RNA, Messenger/metabolism , Racemases and Epimerases/genetics , Up-Regulation , Xenograft Model Antitumor Assays
20.
J Clin Pathol ; 65(4): 309-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22267983

ABSTRACT

AIMS: Increased ß-oxidation of branched-chain fatty acids provides an additional metabolic advantage for cancer cells thereby enhancing tumour development and progression. Alpha-methylacyl coenzyme A racemase (AMACR) is an enzyme essential for the catabolism of branched-chain fatty acids that allows their subsequent ß-oxidation and thus plays an important role in generating biological energy. However, the expression of AMACR has never been systemically investigated in gallbladder carcinoma. This study evaluated the expression status, associations with clinicopathological variables and prognostic implications of AMACR in a well-defined cohort of gallbladder carcinoma and confirmed their expression status in gallbladder carcinoma cells. METHODS: AMACR immunostaining was assessable in 89 cases on tissue microarrays of gallbladder carcinoma, and it was correlated with clinicopathological factors and patient survival. In three gallbladder carcinoma cell lines and one non-tumorigenic cholangiocyte, AMACR mRNA expression was measured by real-time reverse transcription PCR and the endogenous expression of AMACR protein was analysed by western blotting. RESULTS: AMACR overexpression was significantly associated with an advanced primary tumour status (p=0.027) and American Joint Committee on Cancer stage (p=0.027), an increased histological grade (p=0.002) and vascular invasion (p=0.017). Importantly, AMACR overexpression independently predicted worse disease-specific survival (p=0.0452, RR 1.887). Expression levels of AMACR mRNA and total protein in various cells were comparable. The abundance of AMACR expression increased in tumour cells and was even higher in the metastatic cell line. CONCLUSIONS: In primary gallbladder carcinoma, AMACR overexpression was correlated with important prognosticators and independently portended worse outcomes, highlighting the potential prognostic and therapeutic utility of AMACR in gallbladder carcinoma.


Subject(s)
Biomarkers, Tumor/metabolism , Gallbladder Neoplasms/enzymology , Gallbladder/enzymology , Racemases and Epimerases/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Female , Gallbladder Neoplasms/mortality , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...