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










Publication year range
1.
Clin Lab ; 69(7)2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37436392

ABSTRACT

BACKGROUND: Hemoglobin Variant (HBB:c.155 C>A) is a rare mutation caused by ß-globin gene mutation called Hemoglobin North Manchester. So far, its existence has no adverse effect on human body, and it is a rare benign hemoglobin variant. METHODS: We reported a 32-year-old pregnant woman with discordant HbA1c and glucose measurements. In 75 g oral glucose tolerance test (OGTT), the pregnant woman got hyperglycemia at 1h-OGTT and 2h-OGTT. However, the pregnant woman had a low HbA1c of 3.9%. Subsequently, gene sequencing identified a rare mutation in the gene (HBB:c.155 C>A). RESULTS: We report for the first time that a case of North Manchester mutation in a Chinese female patient. In this case, it was found that the North Manchester variant could affect the examination of HbA1c when measured by ion-exchange high-performance liquid chromatography (HPLC), causing in falsely low HbA1c. CONCLUSIONS: Hemoglobin variants may lead to false HbA1c measurement. Clinicians should consider hemoglobin variants when HbA1c results are inconsistent with other laboratory tests.


Subject(s)
Hemoglobins, Abnormal , Adult , Female , Humans , Pregnancy , Chromatography, High Pressure Liquid/methods , East Asian People , Glycated Hemoglobin/genetics , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/analysis , Mutation
2.
Oncol Rep ; 47(4)2022 Apr.
Article in English | MEDLINE | ID: mdl-35211759

ABSTRACT

Cisplatin is one of the most effective chemotherapy drugs for ovarian cancer, but resistance is common. The initial response to platinum­based chemotherapy is as high as 80%, but in most advanced patients, final relapse and death are caused by acquired drug resistance. The development of resistance to therapy in ovarian cancer is a significant hindrance to therapeutic efficacy. The resistance of ovarian cancer cells to chemotherapeutic mechanisms is rather complex and includes multidrug resistance, DNA damage repair, cell metabolism, oxidative stress, cell cycle regulation, cancer stem cells, immunity, apoptotic pathways, autophagy and abnormal signaling pathways. The present review provided an update of recent developments in our understanding of the mechanisms of ovarian cancer platinum­based chemotherapy resistance, discussed current and emerging approaches for targeting these patients and presented challenges associated with these approaches, with a focus on development and overcoming resistance.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism
3.
Medicine (Baltimore) ; 100(39): e27380, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34596157

ABSTRACT

ABSTRACT: Various disease severity scoring systems were currently used in critically ill patients with acute respiratory failure, while their performances were not well investigated.The study aimed to investigate the difference in prognosis predictive value of 4 different disease severity scoring systems in patients with acute respiratory failure.With a retrospective cohort study design, adult patients admitted to intensive care unit (ICU) with acute respiratory failure were screened and relevant data were extracted from an open-access American intensive care database to calculate the following disease severity scores on ICU admission: acute physiology score (APS) III, Sequential Organ Failure Assessment score (SOFA), quick SOFA (qSOFA), and Oxford Acute Severity of Illness Score (OASIS). Hospital mortality was chosen as the primary outcome. Multivariable logistic regression analyses were performed to analyze the association of each scoring system with the outcome. Receiver operating characteristic curve analyses were conducted to evaluate the prognosis predictive performance of each scoring system.A total of 4828 patients with acute respiratory failure were enrolled with a hospital mortality rate of 16.78%. APS III (odds ratio [OR] 1.03, 95% confidence interval [CI] 1.02-1.03), SOFA (OR 1.15, 95% CI 1.12-1.18), qSOFA (OR 1.26, 95% CI 1.11-1.42), and OASIS (OR 1.06, 95% CI 1.05-1.08) were all significantly associated with hospital mortality after adjustment for age and comorbidities. Receiver operating characteristic analyses showed that APS III had the highest area under the curve (AUC) (0.703, 95% CI 0.683-0.722), and SOFA and OASIS shared similar predictive performance (area under the curve 0.653 [95% CI 0.631-0.675] and 0.664 [95% CI 0.644-0.685], respectively), while qSOFA had the worst predictive performance for predicting hospital mortality (0.553, 95% CI 0.535-0.572).These results suggested the prognosis predictive value varied among the 4 different disease severity scores for patients admitted to ICU with acute respiratory failure.


Subject(s)
Hospital Mortality , Intensive Care Units/statistics & numerical data , Respiratory Insufficiency/mortality , Severity of Illness Index , Aged , Aged, 80 and over , Databases, Factual , Female , Humans , Male , Middle Aged , ROC Curve , Retrospective Studies
4.
Am J Transl Res ; 9(1): 103-114, 2017.
Article in English | MEDLINE | ID: mdl-28123637

ABSTRACT

miR-34a is an important molecule that can inhibit the tumor growth. This study aimed to investigate the functional role of miR-34a in hepatocellular carcinoma (HCC) and explore the interaction between miR-34a and histone deacetylase 1 (HDAC1). RT-qPCR was employed to detect the mRNA expression of miR-34a and HDAC1 in 60 HCC tissues. Results showed miR-34a expression in HCC tissues was significantly lower than in normal tissues (P<0.05), but HDAC1 expression in HCC tissues was markedly higher than in normal tissues (P<0.05). In addition, miR-34a expression was negatively related to HDAC1 expression. miR-34a mimic was transfected into HCC cell lines (HepB3 and HepG2). CCK8 assay, colony formation assay and flow cytometry showed miR-34a over-expression could inhibit the proliferation of HCC cells and induce their apoptosis. Western blotting indicated miR-34a over-expression down-regulated the expression of Bcl-2, procaspase-3, procaspase-9 and c-Myc, but up-regulate p21 expression. Bioinformatics analysis indicated HDAC1 was a target gene of miR-34a. Dual Luciferase Reporter Gene Assay and retrieval assay showed miR-34a could act at the 3'UTR of HDAC1 gene to regulate its expression. Thus, miR-34a may inhibit the proliferation of HCC cells and induce their apoptosis via regulating HDAC1 expression. Our findings provide evidence for the diagnosis and therapeutic target of HCC.

6.
Am J Transl Res ; 8(2): 1037-46, 2016.
Article in English | MEDLINE | ID: mdl-27158389

ABSTRACT

MicroRNAs (miRNA) play important regulatory roles in the occurrence and development of cancers. This study aimed to investigate the effects of miR-26a on the proliferation and apoptosis of ovarian cancer cells and explore the potential mechanism. qRT-PCR was performed to measure the miR-26a expression in 46 ovarian cancer tissues, and results showed miR-26a expression reduced significantly when compared with normal ovarian tissues (P<0.05). Moreover, miR-26a expression was related to the extent of cell differentiation and clinical stage of ovarian cancer (P<0.05). miR-26a mimic was transfected into SKOV3 cells and ES2 cells, and CCK8 assay, colony formation assay and flow cytometry showed miR-26a over-expression could significantly inhibit the proliferation of ovarian cancer cells and induce their apoptosis. Bioinformatics analysis revealed Cdc6 was a target gene of miR-26a. dual-luciferase assay and validation assay showed miR-26a could act on the 3'UTR of Cdc6 to regulate Cdc6 expression. These findings suggest that miR-26a may act on the 3'UTR of Cdc6 to regulate Cdc6 expression, which then inhibit the proliferation of ovarian cancer cells and induce their apoptosis. Our findings provide a new target for the diagnosis and therapy of ovarian cancer.

7.
Int J Clin Exp Med ; 8(8): 13388-92, 2015.
Article in English | MEDLINE | ID: mdl-26550271

ABSTRACT

OBJECTIVE: This study aimed to investigate expression level of FOXC2 and its relationship to clinical pathological features of renal cell carcinoma (RCC). METHODS: The expression levels of FOXC2 in RCC tissues and normal renal tissues (62 samples, respectively) were detected by immunohistochemistry and PCR Array. Statistics analyses were done with SPSS to compare the differences between RCC tissues and normal renal tissue, and to explore the relationship between the expression level of FOXC2 and the clinical pathological features of RCC. RESULTS: Expression level of FOXC2 in RCC tissues was significantly higher than in normal renal tissues, and other related cancer genes also highly expressed in RCC tissues. FOXC2 expression was positively associated with clinical stage and pathological grade (P < 0.05), but not significantly related to the gender and age (P > 0.05). CONCLUSION: The expression of FOXC2 in renal cell carcinoma was significantly higher than that in normal renal tissues. It is suggested that FOXC2 might play a crucial role in the occurrence and development of RCC and could be an important prognostic indicator for clinical therapy.

8.
PLoS One ; 7(4): e34265, 2012.
Article in English | MEDLINE | ID: mdl-22496786

ABSTRACT

Histone deacetylases (HDACs) are known to play a central role in the regulation of several cellular properties interlinked with the development and progression of cancer. Recently, HDAC1 has been reported to be overexpressed in hepatocellular carcinoma (HCC), but its biological roles in hepatocarcinogenesis remain to be elucidated. In this study, we demonstrated overexpression of HDAC1 in a subset of human HCCs and liver cancer cell lines. HDAC1 inactivation resulted in regression of tumor cell growth and activation of caspase-independent autophagic cell death, via LC3B-II activation pathway in Hep3B cells. In cell cycle regulation, HDAC1 inactivation selectively induced both p21(WAF1/Cip1) and p27(Kip1) expressions, and simultaneously suppressed the expression of cyclin D1 and CDK2. Consequently, HDAC1 inactivation led to the hypophosphorylation of pRb in G1/S transition, and thereby inactivated E2F/DP1 transcription activity. In addition, we demonstrated that HDAC1 suppresses p21(WAF1/Cip1) transcriptional activity through Sp1-binding sites in the p21(WAF1/Cip1) promoter. Furthermore, sustained suppression of HDAC1 attenuated in vitro colony formation and in vivo tumor growth in a mouse xenograft model. Taken together, we suggest the aberrant regulation of HDAC1 in HCC and its epigenetic regulation of gene transcription of autophagy and cell cycle components. Overexpression of HDAC1 may play a pivotal role through the systemic regulation of mitotic effectors in the development of HCC, providing a particularly relevant potential target in cancer therapy.


Subject(s)
Autophagy , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Liver Neoplasms/pathology , Mitosis/physiology , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Colony-Forming Units Assay , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Liver/cytology , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Luciferases/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Retinoblastoma Protein , Sp1 Transcription Factor , Transcriptional Activation
9.
J Cell Biochem ; 113(6): 2167-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492270

ABSTRACT

Histone deacetylase 2 (HDAC2) is crucial for embryonic development, affects cytokine signaling relevant for immune responses, and is often significantly overexpressed in solid tumors, but little is known of its role in human lung cancer. In this study, we demonstrated the aberrant expression of HDAC2 in lung cancer tissues and investigated oncogenic properties of HDAC2 in human lung cancer cell lines. HDAC2 inactivation resulted in regression of tumor cell growth and activation of cellular apoptosis via p53 and Bax activation and Bcl2 suppression. In cell cycle regulation, HDAC2 inactivation caused induction of p21WAF1/CIP1 expression, and simultaneously suppressed the expressions of cyclin E2, cyclin D1, and CDK2, respectively. Consequently, this led to the hypophosphorylation of pRb protein in G1/S transition and thereby inactivated E2F/DP1 target gene transcriptions of A549 cells. In addition, we demonstrated that HDAC2 directly regulated p21WAF1/CIP1 expression in a p53-independent manner. However, HDAC1 was not related to p21WAF1/CIP1 expression and tumorigenesis of lung cancer. Lastly, we observed that sustained-suppression of HDAC2 in A549 lung cancer cells attenuated in vitro tumorigenic properties and in vivo tumor growth of the mouse xenograft model. Taken together, we suggest that the aberrant regulation of HDAC2 and its epigenetic regulation of gene transcription in apoptosis and cell cycle components play an important role in the development of lung cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cell Cycle Proteins/metabolism , Histone Deacetylase 2/metabolism , Lung Neoplasms/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , E2F Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Phosphorylation , RNA Interference , RNA, Small Interfering , Retinoblastoma Protein/metabolism , Transcription, Genetic , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/metabolism
10.
PLoS One ; 6(11): e28103, 2011.
Article in English | MEDLINE | ID: mdl-22132221

ABSTRACT

Histone deacetylase 2 (HDAC2) is crucial for embryonic development, affects cytokine signaling relevant for immune responses and is often significantly overexpressed in solid tumors; but little is known about its role in human hepatocellular carcinoma (HCC). In this study, we showed that targeted-disruption of HDAC2 resulted in reduction of both tumor cell growth and de novo DNA synthesis in Hep3B cells. We then demonstrated that HDAC2 regulated cell cycle and that disruption of HDAC2 caused G1/S arrest in cell cycle. In G1/S transition, targeted-disruption of HDAC2 selectively induced the expression of p16(INK4A) and p21(WAF1/Cip1), and simultaneously suppressed the expression of cyclin D1, CDK4 and CDK2. Consequently, HDAC2 inhibition led to the down-regulation of E2F/DP1 target genes through a reduction in phosphorylation status of pRb protein. In addition, sustained suppression of HDAC2 attenuated in vitro colony formation and in vivo tumor growth in a mouse xenograft model. Further, we found that HDAC2 suppresses p21(WAF1/Cip1) transcriptional activity via Sp1-binding site enriched proximal region of p21(WAF1/Cip1) promoter. In conclusion, we suggest that the aberrant regulation of HDAC2 may play a pivotal role in the development of HCC through its regulation of cell cycle components at the transcription level providing HDAC2 as a relevant target in liver cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , G1 Phase/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylase 2/metabolism , S Phase , Animals , Binding Sites , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enzyme Activation/drug effects , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Targeting , Histone Deacetylase Inhibitors/pharmacology , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mitogens/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/metabolism , S Phase/drug effects , S Phase/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects , Wnt Signaling Pathway/drug effects
11.
Toxicol Appl Pharmacol ; 250(2): 162-9, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20955722

ABSTRACT

In a previous report we demonstrated that the transcriptomic response of liver tissue was specific to toxicants, and a characteristic molecular signature could be used as an early prognostic biomarker in rats. It is necessary to determine the transcriptomic response to toxicants in peripheral blood for application to the human system. Volatile organic compounds (VOCs) comprise a major group of pollutants which significantly affect the chemistry of the atmosphere and human health. In this study we identified and validated the specific molecular signatures of toxicants in rat whole blood as early predictors of environmental toxicants. VOCs (dichloromethane, ethylbenzene, and trichloroethylene) were administered to 11-week-old SD male rats after 48h of exposure, peripheral whole blood was subjected to expression profiling analysis. Unsupervised gene expression analysis resulted in a characteristic molecular signature for each toxicant, and supervised analysis identified 1,217 outlier genes as distinct molecular signatures discerning VOC exposure from healthy controls. Further analysis of multi-classification suggested 337 genes as early detective molecular markers for three VOCs with 100% accuracy. A large-scale gene expression analysis of a different VOC exposure animal model suggested that characteristic expression profiles exist in blood cells and multi-classification of this VOC-specific molecular signature can discriminate each toxicant at an early exposure time. This blood expression signature can thus be used as discernable surrogate marker for detection of biological responses to VOC exposure in an environment.


Subject(s)
Benzene Derivatives/toxicity , Gene Expression Regulation/drug effects , Methylene Chloride/toxicity , Trichloroethylene/toxicity , Animals , Biomarkers/metabolism , Environmental Exposure/adverse effects , Gene Expression Profiling , Male , Rats , Rats, Sprague-Dawley , Time Factors
12.
Oncol Rep ; 24(3): 607-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664964

ABSTRACT

Gastric carcinoma is the most common neoplasm in Southeast Asian populations and is the second leading cause of cancer death worldwide. Annexins are a family of cytosolic calcium and membrane binding proteins that have been implicated in a wide variety of cell functions. Recent studies have suggested that Annexin A10 (ANXA10), a member of the Annexin protein family, is down-regulated in specific types of cancer. However, the underlying molecular mechanisms of the dysregulation of ANXA10 remain to be elucidated. In the present study, to investigate the biological effects of ANXA10 on gastric carcinoma, aberrant expression of ANXA10 was evaluated by Western blot analysis, reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC), in gastric cancer tissues and cell lines. Decreased expression of ANXA10 was observed in five selected gastric cancer tissues compared to the normal surrounding mucosa. In the cancer cell lines, seven out of nine selected gastric cancer cell lines had no detectable ANXA10 by RT-PCR. Among these, when an ANXA10 expressing plasmid was introduced into MKN-1 cells, cell growth was suppressed and apoptosis augmented. The results of this study demonstrated that ANXA10 was aberrantly regulated in gastric carcinoma and suggests that down-regulation of ANXA10 might be involved in gastric carcinogenesis. In addition, ANXA10 may play a role, as a tumor suppressor, in the development and progression of gastric cancer.


Subject(s)
Annexins/metabolism , Carcinoma/metabolism , Cell Proliferation , Stomach Neoplasms/metabolism , Annexins/genetics , Apoptosis , Blotting, Western , Carcinoma/genetics , Carcinoma/pathology , Case-Control Studies , Cell Line, Tumor , Down-Regulation , Humans , Immunohistochemistry , Republic of Korea , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transfection , Up-Regulation
13.
J Cell Biochem ; 110(3): 687-96, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20512928

ABSTRACT

The Wnt/beta-catenin signaling pathway regulates various aspects of development and plays important role in human carcinogenesis. Nemo-like kinase (NLK), which is mediator of Wnt/beta-catenin signaling pathway, phosphorylates T-cell factor/lymphoid enhancer factor (TCF/LEF) factor and inhibits interaction of beta-catenin/TCF complex. Although, NLK is known to be a tumor suppressor in Wnt/beta-catenin signaling pathway of colon cancer, the other events occurring downstream of NLK pathways in other types of cancer remain unclear. In the present study, we identified that expression of NLK was significantly up-regulated in the HCCs compared to corresponding normal tissues in five selected tissue samples. Immunohistochemical analysis showed significant over-expression of NLK in the HCCs. Targeted-disruption of NLK suppressed cell growth and arrested cell cycle transition. Suppression of NLK elicited anti-mitogenic properties of the Hep3B cells by simultaneous inhibition of cyclinD1 and CDK2. The results of this study suggest that NLK is aberrantly regulated in HCC, which might contribute to the mitogenic potential of tumor cells during the initiation and progression of hepatocellular carcinoma; this process appears to involve the induction of CDK2 and cyclin D1 and might provide a novel target for therapeutic intervention in patients with liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Liver Neoplasms/metabolism , Transcription Factor TFIIIA/metabolism , Apoptosis/physiology , Blotting, Western , Carcinoma, Hepatocellular/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Cell Proliferation , Cyclin D1/genetics , Cyclin-Dependent Kinase 2/genetics , Gene Expression , Gene Expression Regulation , Gene Silencing , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Membrane Transport Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcription Factor TFIIIA/genetics
14.
Int J Oncol ; 35(6): 1257-64, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19885547

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer death worldwide. The number of cases of HCC has continued to increase in recent decades. Previous studies have suggested that S100P, a member of the S100P calcium-binding protein family, is aberrantly regulated in several malignant neoplasms. However, the underlying molecular mechanisms of the dysregulation of S100P remain to be elucidated. To investigate biological effects of S100P on hepatocarcinogenesis, aberrant expression of S100P was investigated by immunohistochemistry (IHC), Western blot analysis and reverse transcriptase-polymerase chain reaction (RT-PCR) in HCC tissues and cell lines. Endogenous expression of S100P was disrupted by the RNA interference-mediated protein knockdown method in the human Hep3B liver cancer cell line. Then, cell growth and cellular apoptosis were compared with control siRNA transfectants. The effects of S100P-silencing on the major components of cell cycle regulation were assessed by Western blot analysis. As results, elevated levels of S100P were observed in the HCC tissues compared to the corresponding normal tissues. Targeted disruption of S100P suppressed cell growth and augmented cellular apoptosis. In addition, inhibition of S100P resulted in the down-regulation of cyclinD1 and CDK2. In conclusion, this study showed over-expression of S100P in HCC. The aberrant regulation of S100P in HCC might activate cyclin D1 and CDK expression and contribute to the mitogenic potential of tumor cells during HCC carcinogenesis. These findings provide information that suggests new therapeutic strategies for the treatment of liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carrier Proteins/genetics , Cell Proliferation , Cyclin D1/genetics , Cyclin-Dependent Kinase 2/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Nuclear Proteins/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
15.
Oncol Rep ; 22(3): 475-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639191

ABSTRACT

The transforming growth factor receptor III (TGFbetaRIII) is the most abundant and essential TGF-beta binding protein that functions as a co-receptor with other receptors in TGF-beta signaling. In earlier studies, expression of TGFbetaRIII was reported to be decreased in a variety of human cancers. Functional assessment of TGFbetaRIII was performed in many previously studied cancers but not in hepatocellular carcinoma. Therefore, in this study, we investigated the expression and genetic alterations of TGFbetaRIII in hepatocellular carcinoma (HCC) by quantitative real-time PCR (qRT-PCR) and single-strand conformation polymorphism (SSCP) analysis. The qRT-PCR showed down-regulation of TGFbetaRIII in the tumor samples. To investigate whether genetic alterations mediated decreased expression of TGFbetaRIII, we performed mutation analysis of 67 human HCC tissues by SSCP and direct sequencing. We found five previously reported and one novel single nucleotide polymorphisms in exons 2, 3, 5, 13 and 14, but no mutations were detected. These polymorphisms were not associated with amino acid changes except for a base change found in exon 2 (TCC-->TTC, S15F). The loss of heterozygosity (LOH) analysis performed on 10 tumors and corresponding normal pairs, showed a low rate of LOH (2/10). The results of this study suggest that TGFbetaRIII is transcriptionally down-regulated in hepatocellular carcinoma. In addition, genetic alterations did not appear to be associated with the reduced expression level of TGFbetaRIII. To clarify the role of TGFbetaRIII in hepatocellular tumor development and progression, functional analysis is needed in future studies.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Loss of Heterozygosity , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Down-Regulation , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Signal Transduction , Transforming Growth Factor beta/physiology
16.
Int J Mol Med ; 24(2): 205-26, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578794

ABSTRACT

Histone deacetylase (HDAC) inhibitors are emerging as an exciting new class of potential anti-cancer agents for the treatment of solid and hematological malignancies. However, the best characterized HDAC function concerns the control of gene expression via the regulation of transcription activation or repression. To understand the genome-wide effects of HDAC inhibition on gene regulation, we performed serial gene expression analyses from 0 to 48 h after treating MDA-MB-435, a melanoma-derived highly metastatic tumor cell line, with Apicidin, a HDAC inhibitor. Combined-transcriptomic analysis of large-scale molecular changes induced by Apicidin resulted in the identification of 631 outlier genes that were continuously up- or down-regulated during the 48 h study period. When the 631 outlier genes were mapped to known biological processes, cell-cycle suppression emerged as the function most elicited by Apicidin. In addition comprehensive negative cell-cycle regulation by Apicidin was dissected using gene expression data and validated by Western blot analysis. We suggest the 631 outlier genes as a characteristic molecular signature for Apicidin, and propose concurrent transcriptional suppression of major components of cell-cycle regulatory circuit as potent anti-tumor mechanism of Apicidin. Genetic elements identified during this study also provide the possibility of novel therapeutic interventions in tumor metastasis.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Histone Deacetylase Inhibitors , Peptides, Cyclic/pharmacology , Apoptosis/genetics , Blotting, Western , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Humans , Models, Biological , Oligonucleotide Array Sequence Analysis , Smad3 Protein/genetics , Smad3 Protein/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...