ABSTRACT
INTRODUCTION AND OBJECTIVES: Hepatocellular carcinoma (HCC) is the second most lethal cancer around the world, with poor survival rate and high metastasis rate in patients. Long noncoding RNAs (lncRNAs) have been reported to modulate the initiation and development of liver cancer. We aimed to investigate the role of lncRNA MAGI2-AS3 in HCC and underlying mechanisms. MATERIALS AND METHODS: The expression levels of MAGI2-AS3 in plasma of HCC patients and the control participants were measured by qPCR. Hep3B and MHCC97-H cells were transfected with MAGI2-AS3 and ROCK2 expression vectors. Cell migration and invasion of HCC cells transfected with the vectors were investigated by transwell assay. In addition, flow cytometry and western blot were performed for apoptosis detection. RESULTS: We found that MAGI2-AS3 was downregulated in plasma of early stage HCC patients compared to healthy controls. After surgical resection, the expression levels of MAGI2-AS3 were increased compared to pretreatment levels on the day of discharge. During the follow-up, MAGI2-AS3 was downregulated in patients developed distant recurrence, but not in other patients compared to the levels measured on the day of discharge. In HCC cells, overexpression of MAGI2-AS3 mediated the downregulation of ROCK2. Cell invasion and migration assay showed that overexpression of MAGI2-AS3 mediated the decreased cell invasion and migration rate, while ROCK2 played an opposite role and attenuated the effects of overexpression of MAGI2-AS3. CONCLUSION: Our study indicated that MAGI2-AS3 was downregulated in the distant recurrence of HCC after surgical resection and affected the invasion and migration of HCC cells via ROCK2.
Subject(s)
Carcinoma, Hepatocellular/surgery , Cell Movement , Hepatectomy/adverse effects , Liver Neoplasms/surgery , RNA, Long Noncoding/metabolism , rho-Associated Kinases/metabolism , Adult , Aged , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Case-Control Studies , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , RNA, Long Noncoding/genetics , Signal Transduction , Treatment Outcome , rho-Associated Kinases/geneticsABSTRACT
BACKGROUND & AIMS: A causal link has recently been established between epigenetic alterations and hepatocarcinogenesis, indicating that epigenetic inhibition may have therapeutic potential. We aimed to identify and target epigenetic modifiers that show molecular alterations in hepatocellular carcinoma (HCC). METHODS: We studied the molecular-clinical correlations of epigenetic modifiers including bromodomains, histone acetyltransferases, lysine methyltransferases and lysine demethylases in HCC using The Cancer Genome Atlas (TCGA) data of 365 patients with HCC. The therapeutic potential of epigenetic inhibitors was evaluated in vitro and in vivo. RNA sequencing analysis and its correlation with expression and clinical data in the TCGA dataset were used to identify expression programs normalized by Jumonji lysine demethylase (JmjC) inhibitors. RESULTS: Genetic alterations, aberrant expression, and correlation between tumor expression and poor patient prognosis of epigenetic enzymes are common events in HCC. Epigenetic inhibitors that target bromodomain (JQ-1), lysine methyltransferases (BIX-1294 and LLY-507) and JmjC lysine demethylases (JIB-04, GSK-J4 and SD-70) reduce HCC aggressiveness. The pan-JmjC inhibitor JIB-04 had a potent antitumor effect in tumor bearing mice. HCC cells treated with JmjC inhibitors showed overlapping changes in expression programs related with inhibition of cell proliferation and induction of cell death. JmjC inhibition reverses an aggressive HCC gene expression program that is also altered in patients with HCC. Several genes downregulated by JmjC inhibitors are highly expressed in tumor vs. non-tumor parenchyma, and their high expression correlates with a poor prognosis. We identified and validated a 4-gene expression prognostic signature consisting of CENPA, KIF20A, PLK1, and NCAPG. CONCLUSIONS: The epigenetic alterations identified in HCC can be used to predict prognosis and to define a subgroup of high-risk patients that would potentially benefit from JmjC inhibitor therapy. LAY SUMMARY: In this study, we found that mutations and changes in expression of epigenetic modifiers are common events in human hepatocellular carcinoma, leading to an aggressive gene expression program and poor clinical prognosis. The transcriptional program can be reversed by pharmacological inhibition of Jumonji enzymes. This inhibition blocks hepatocellular carcinoma progression, providing a novel potential therapeutic strategy.
Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis , Carcinoma, Hepatocellular , Epigenesis, Genetic/drug effects , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Liver Neoplasms , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Centromere Protein A/genetics , Drug Discovery , Humans , Kinesins/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Mice , Mutation , Prognosis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Transcriptome , Polo-Like Kinase 1ABSTRACT
INTRODUCTION AND AIM: Developing reliable biomarkers for hepatocellular carcinoma (HCC) patients who are at a high risk of recurrence after curative hepatic resection is very important for determining subsequent therapeutic strategies. We investigated the role of the cell cycle factor NIMA-related kinase 2 (NEK2) in HCC progression in hepatoma cells and post-surgery patients. MATERIAL AND METHODS: The effects of NEK2 on proliferation, invasion and migration of hepatoma HuH7 and SK-Hep1 cells were evaluated. In a post-surgery HCC cohort (N = 97), the Nek2 induction levels in the tumors were examined with real-time RT-PCR analysis, and the results were analyzed for their correlations with recurrence. RESULTS: NEK2 promoted G1 to S phase cell cycle progression by causing increases in cyclin D1 and AKT phosphorylation and decreases in the cyclin-dependent kinase inhibitor p27, indicating that NEK2 plays an important role during interphase in addition to its previously identified role in M phase. NEK2 also enhanced the proliferation, migration and invasion of hepatoma cells and regulated the expression of E-cadherin and MMP9. The Nek2 mRNA levels in the tumors were highly correlated with recurrence rates in the post-surgery HCC patients. Combined evaluation of the tumor AJCC stage and the Nek2 level can serve as a reliable method for predicting the relative risk of HCC recurrence in these patients. CONCLUSIONS: NEK2 plays a significant role in cell cycle progression in the inter- and M-phases. NEK2 enhances HCC metastasis and is correlated with recurrence and thus can potentially serve a promising high-risk biomarker for HCC.
Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/surgery , Hepatectomy/adverse effects , Liver Neoplasms/surgery , NIMA-Related Kinases/metabolism , Neoplasm Recurrence, Local , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Biomarkers, Tumor/genetics , Cadherins/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , NIMA-Related Kinases/genetics , Neoplasm Invasiveness , Neoplasm Staging , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Risk Factors , Signal Transduction , Time Factors , Treatment OutcomeABSTRACT
BACKGROUND: Genistein (5,7-Dihydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) is the most abundant isoflavone in soybean, which has been associated with a lower risk of development of cancer and cardiovascular diseases. Of particular interest regarding cancer preventive properties of flavonoids is their interaction with cytochrome P450 enzymes (CYPs). However, contradictory data report the effect of genistein on expression of СYPs enzymes. OBJECTIVE: The aim of this study was to investigate the effects of genistein on cytochrome P450 (CYP) gene expression levels in human hepatocellular carcinoma (HepG2/C3A) and colon adenocarcinoma (HT29) cells. METHODS: Real-time RT-PCR was used to examine the expression of genes families involved in xenobiotic metabolism, such as CYP1 (CYP1A1, CYP1B1), CYP2 (CYP2E1, CYP2D6), CYP3 (CYP3A4); and of a family involved in the catabolism of the all-trans-retinoic acid (ATRA), CYP26 (CYP26A1, CYP26B1). RESULTS: RT-qPCR data analysis showed that after 12 h of exposure of HepG2/C3A cells to genistein (5 and 50 µM) there was an upregulation of CYP1A1 and CYP1B1 and downregulation of CYP2D6, CYP26A1 and CYP26B1 mRNA levels. There was no change in the mRNA levels of CYP P450 genes in HT29 cells. CONCLUSION: Our results suggest that treatment with genistein in non-toxic concentrations may impact the expression level of CYPs involved in the biotransformation of xenobiotics and drug metabolizing enzymes. Moreover, the downregulation of ATRA metabolism-related genes opens a new research path for the study of genistein as retinoic acid metabolism blocking agent for treating cancer and other pathologies.
Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Genistein/pharmacology , Liver Neoplasms/enzymology , Biotransformation , Carcinoma, Hepatocellular/genetics , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , HT29 Cells , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Xenobiotics/metabolismABSTRACT
BACKGROUND: The intrinsic heterogeneity of hepatocellular carcinoma (HCC) represents a great challenge for its molecular classification and for detecting predictive biomarkers. Aldo-keto reductase (Akr) family members have shown differential expression in human HCC, while AKR1B10 overexpression is considered a biomarker; AKR7A3 expression is frequently reduced in HCC. AIMS: To investigate the time-course expression of Akr members in the experimental hepatocarcinogenesis. METHODS: Using DNA-microarray data, we analyzed the time-course gene expression profile from nodules to tumors (4-17 months) of 17 Akr members induced by the resistant hepatocyte carcinogenesis model in the rat. RESULTS: The expression of six members (Akr1c19, Akr1b10, Akr7a3, Akr1b1, Akr1cl1, and Akr1b8) was increased, comparable to that of Ggt and Gstp1, two well-known liver cancer markers. In particular, Akr7a3 and Akr1b10 expression also showed a time-dependent increment at mRNA and protein levels in a second hepatocarcinogenesis model induced with diethylnitrosamine. We confirmed that aldo-keto reductases 7A3 and 1B10 were co-expressed in nine biopsies of human HCC, independently from the presence of glypican-3 and cytokeratin-19, two well-known HCC biomarkers. Because it has been suggested that expression of Akr members is regulated through NRF2 activity at the antioxidant response element (ARE) sequences, we searched and identified at least two ARE sites in Akr1b1, Akr1b10, and Akr7a3 from rat and human gene sequences. Moreover, we observed higher NRF2 nuclear translocation in tumors as compared with non-tumor tissues. CONCLUSIONS: Our results demonstrate that Akr7a3 mRNA and protein levels are consistently co-expressed along with Akr1b10, in both experimental liver carcinogenesis and some human HCC samples. These results highlight the presence of AKR7A3 and AKR1B10 from early stages of the experimental HCC and introduce them as a potential application for early diagnosis, staging, and prognosis in human cancer.
Subject(s)
Aldehyde Reductase/metabolism , Aldo-Keto Reductase Family 1 member B10/metabolism , Aldo-Keto Reductases/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Aldehyde Reductase/genetics , Aldo-Keto Reductase Family 1 member B10/genetics , Aldo-Keto Reductases/genetics , Animals , Biomarkers/metabolism , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Humans , Liver Neoplasms/etiology , Liver Neoplasms/pathology , RNA, Messenger/metabolism , Rats, Inbred F344ABSTRACT
In cancers, apoptosis signaling pathways and cell survival and growth pathways responsible for resistance to conventional treatments, such as Pi3K/Akt/mTOR and mitogen-activated protein kinase (MAPK) become dysregulated. Recently, alternative treatments to promote tumor cell death have become important. The present study reports on the antitumor and cytoprotective action of gold nanoparticles (GNPs) and carvedilol in combination and in isolated application. Apoptosis was analyzed by FITC/propidium iodide staining flow cytometry; caspase-3, caspase-8, Bcl-2 and MAPK/ERK activity by immunofluorescence microscopy; gene expression of proteins related to cell death as Akt, mTOR, EGFR, MDR1, survivin, FADD and Apaf, by the real-time PCR; and western blot analysis for MAPK/ERK, Akt and mTOR. Oxidative stress evaluation was performed by reduced glutathione (GSH) and malondialdehyde (MDA) levels. Intracellular GNPs targets were identified by transmission electron microscopy. After exposure to a combination of GNPs (6.25 µg/ml) and carvedilol (3 µM), death as promoted by apoptosis was detected using flow cytometry, for expression of pro-apoptotic proteins FADD, caspase-3, caspase-8 and sub-regulation of anti-apoptotic MAPK/ERK, Akt, mTOR, EGFR and MDR1 resistance. Non-tumor cell cytoprotection with GSH elevation and MDA reduction levels was detected. GNPs were identified within the cell near to the nucleus when combined with carvedilol. The combination of GNP and carvedilol promoted downregulation of anti-apoptotic and drug resistance genes, over-regulation of pro-apoptotic proteins in tumor cells, as well as cytoprotection of non-tumor cells with reduction of apoptosis and oxidative stress.
Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carbazoles/pharmacology , Gold/administration & dosage , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Metal Nanoparticles/administration & dosage , Propanolamines/pharmacology , Apoptosis/drug effects , Carbazoles/administration & dosage , Carvedilol , ErbB Receptors/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Oxidative Stress/drug effects , Propanolamines/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolismABSTRACT
Telomeres are repetitive DNA sequences at linear chromosome termini, protecting chromosomes against end-to-end fusion and damage, providing chromosomal stability. Telomeres shorten with mitotic cellular division, but are maintained in cells with high proliferative capacity by telomerase. Loss-of-function mutations in telomere-maintenance genes are genetic risk factors for cirrhosis development in humans and murine models. Telomerase deficiency provokes accelerated telomere shortening and dysfunction, facilitating genomic instability and oncogenesis. Here we examined whether telomerase mutations and telomere shortening were associated with hepatocellular carcinoma (HCC) secondary to cirrhosis. Telomere length of peripheral blood leukocytes was measured by Southern blot and qPCR in 120 patients with HCC associated with cirrhosis and 261 healthy subjects. HCC patients were screened for telomerase gene variants (in TERT and TERC) by Sanger sequencing. Age-adjusted telomere length was comparable between HCC patients and healthy subjects by both Southern blot and qPCR. Four non-synonymous TERT heterozygous variants were identified in four unrelated patients, resulting in a significantly higher mutation carrier frequency (3.3%) in patients as compared to controls (p = 0.02). Three of the four variants (T726M, A1062T, and V1090M) were previously observed in patients with other telomere diseases (severe aplastic anemia, acute myeloid leukemia, and cirrhosis). A novel TERT variant, A243V, was identified in a 65-year-old male with advanced HCC and cirrhosis secondary to chronic hepatitis C virus (HCV) and alcohol ingestion, but direct assay measurements in vitro did not detect modulation of telomerase enzymatic activity or processivity. In summary, constitutional variants resulting in amino acid changes in the telomerase reverse transcriptase were found in a small proportion of patients with cirrhosis-associated HCC.
Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Liver Cirrhosis/enzymology , Liver Neoplasms/enzymology , Telomerase/metabolism , Telomere/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Male , Middle Aged , Mutation/genetics , Polymerase Chain Reaction , Telomerase/genetics , Young AdultABSTRACT
Dysregulation of mammalian target of rapamycin (mTOR) in hepatocellular carcinoma (HCC) represents a valuable treatment target. Recent studies have developed a highly-selective and potent mTOR kinase inhibitor, CZ415. Here, we showed that nM concentrations of CZ415 efficiently inhibited survival and induced apoptosis in HCC cell lines (HepG2 and Huh-7) and primary-cultured human HCC cells. Meanwhile, CZ415 inhibited proliferation of HCC cells, more potently than mTORC1 inhibitors (rapamycin and RAD001). CZ415 was yet non-cytotoxic to the L02 human hepatocytes. Mechanistic studies showed that CZ415 disrupted assembly of mTOR complex 1 (mTORC1) and mTORC2 in HepG2 cells. Meanwhile, activation of mTORC1 (p-S6K1) and mTORC2 (p-AKT, Ser-473) was almost blocked by CZ415. In vivo studies revealed that oral administration of CZ415 significantly suppressed HepG2 xenograft tumor growth in severe combined immuno-deficient (SCID) mice. Activation of mTORC1/2 was also largely inhibited in CZ415-treated HepG2 tumor tissue. Together, these results show that CZ415 blocks mTORC1/2 activation and efficiently inhibits HCC cell growth in vitro and in vivo.
Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cyclic S-Oxides/pharmacology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclic S-Oxides/chemical synthesis , Cyclic S-Oxides/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, CulturedABSTRACT
Using the conventional Haberkorn approach, it is evaluated the recombination of the radical pair (RP) singlet spin state to study theoretically the cytoprotective effect of an extremely-low-frequency electromagnetic field (ELF-EMF) on early stages of hepatic cancer chemically induced in rats. The proposal is that ELF-EMF modulates the interconversion rate of singlet and triplet spin states of the RP populations modifying the products from the metabolization of carcinogens. Previously, we found that the daily treatment with ELF-EMF 120 Hz inhibited the number and area of preneoplastic lesions in chemical carcinogenesis. The singlet spin population is evaluated diagonalizing the spin density matrix through the Lanczos method in a radical pair mechanism (RPM). Using four values of the interchange energy, we have studied the variations over the singlet population. The low magnetic field effect as a test of the influence over the enzymatic chemical reaction is evaluated calculating the quantum yield. Through a bootstrap technique the range is found for the singlet decay rate for the process. Applying the quantum measurements concept, we addressed the impact toward hepatic cells. The result contributes to improving our understanding of the chemical carcinogenesis process affected by charged particles that damage the DNA.
Subject(s)
Liver Neoplasms/therapy , Magnetic Field Therapy , Models, Biological , Animals , Carcinogenesis/metabolism , Cytochrome P-450 Enzyme System/metabolism , Diethylnitrosamine , Disease Models, Animal , Electromagnetic Fields , Hepatocytes/enzymology , Liver Neoplasms/enzymology , Quantum Theory , Time FactorsABSTRACT
Of late, many studies are attempting to find new molecules with anticancer properties, especially those with the capability to inhibit cell growth. The aim of this work was to evaluate nerolidol, a plant-based compound, as its cytotoxicity, genotoxicity, antiproliferative and apoptotic induction, cell cycle, mitochondrial membrane potential and RT-qPCR of transcripts related to those pathways in the human hepatocellular carcinoma cell line (HepG2/C3A). Only cis-nerolidol (C-NER) demonstrated cytotoxicity (100-250 µM) activity and was selected to conduct the following experiments. C-NER did not show genotoxic activity, but altered the mitochondrial membrane potential, reduced cell proliferation by arresting cell cycle in G1 phase and induced cell death. RT-qPCR showed that C-NER down-regulated genes related to apoptosis (BAK1, BAX, CAPN1, CASP8, CASP9, PARP1 and TP53), cell cycle (CCND1, CCNE1, CDK1 and CDK2), xenobiotic metabolism (CYP2D6 and CYP3A4) and paraptosis (IGF1R receptor). Up-regulation was seen in case of genes related to cell survival (BBC3 and MYC) and reticulum stress protein response (EIF2AK3 and ERN1) and xenobiotic metabolism (CYP1A2 and CYP2C19). We deduced that the antiproliferative activity of C-NER is attributable to its modulation of the cyclins and cyclin-dependent kinases as these proteins are necessary for G1/S phase transition. EIF2AK3, ERN1, CYP2C19 and CYP1A2 up-regulation suggests that endoplasmic reticulum stress was induced owing to the increased activity of cytochrome P450 enzymes. Caspase-independent cell death was also observed, indicating that another type of cell death, paraptosis, was triggered. Our results indicate that C-NER has considerable potential in anticancer therapy because it modulates important molecular targets of cell survival and proliferation.
Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19/metabolism , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Sesquiterpenes/pharmacology , Activation, Metabolic , Antineoplastic Agents, Phytogenic/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , G1 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Oxidation-Reduction , Sesquiterpenes/metabolism , Time FactorsABSTRACT
Background. The protein encoded by PARK2 gene is a component of the ubiquitin-proteasome system that mediates targeting of proteins for the degradation pathway. Genetic variations at PARK2 gene were linked to various diseases including leprosy, typhoid and cancer. The present study investigated the association of single nucleotide polymorphisms (SNPs) in the PARK2 gene with the development of hepatitis C virus (HCV) infection and its progression to severe liver diseases. MATERIAL AND METHODS: A total of 800 subjects, including 400 normal healthy subjects and 400 HCV-infected patients, were analyzed in this study. The patients were classified as chronic HCV patients (group I), patients with cirrhosis (group II) and patients with hepatocellular carcinoma (HCC) in the context of cirrhosis (group III). DNA was extracted and was genotyped for the SNPs rs10945859, rs2803085, rs2276201 and rs1931223. RESULTS: Among these SNPs, CT genotype of rs10945859 was found to have a significant association towards the clinical progression of chronic HCV infection to cirrhosis alone (OR = 1.850; 95% C. I. 1.115-3.069; p = 0.016) or cirrhosis and HCC (OR = 1.768; 95% C. I. 1.090-2.867; p value = 0.020). CONCLUSION: SNP rs10945859 in the PARK2 gene could prove useful in predicting the clinical outcome in HCV-infected patients.
Subject(s)
Carcinoma, Hepatocellular/genetics , Hepatitis C, Chronic/genetics , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Polymorphism, Single Nucleotide , Ubiquitin-Protein Ligases/genetics , Adult , Aged , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/virology , Case-Control Studies , Chi-Square Distribution , Disease Progression , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/enzymology , Hepatitis C, Chronic/virology , Humans , Linkage Disequilibrium , Liver Cirrhosis/diagnosis , Liver Cirrhosis/enzymology , Liver Cirrhosis/virology , Liver Neoplasms/diagnosis , Liver Neoplasms/enzymology , Liver Neoplasms/virology , Male , Middle Aged , Odds Ratio , Phenotype , Risk Factors , Young AdultABSTRACT
Introduction and aim. Hepatocellular carcinoma (HCC) is the most common primary malignant liver tumor. It is primarily caused by hepatic cirrhosis or chronic viral hepatitis. Hepatic carcinogenesis is associated with increased oxidative stress. Thus, the aim of our study was to assess expression of the genes involved in the homeostasis of oxidative stress in patients with HCC. MATERIAL AND METHODS: The study was performed on 32 patients with primary HCC (verified by liver histology in 29 patients) and 27 control subjects (in 11 subjects, liver histology was available either with no or minimal changes in the liver tissue). Gene expressions of heme oxygenase 1 (HMOX1), biliverdin reductase A/B (BLVRA/B), NADPH oxidase 2 (NOX2) and p22phox were analyzed in the liver and peripheral blood leukocytes (PBL) in the subjects. RESULTS: Compared to controls, almost a 3 times higher mRNA level of BLVRA was detected in livers of HCC patients (p = 0.002); while those of BLVRB as well as HMOX1 were unchanged (p > 0.05). In accord with these results in the liver tissue, BLVRA mRNA levels in PBL were also significantly increased in HCC patients (p = 0.012). mRNA levels of NOX2 and p22phox in the liver tissue, although higher in HCC patients, did not differ significantly compared to control subjects (p > 0.05). Nevertheless, NOX2 mRNA level in PBL was significantly higher in HCC patients (p = 0.003). CONCLUSIONS: BLVRA mRNA levels in the liver as well as in PBL are significantly higher in HCC patients most likely as a feedback mechanism to control increased oxidative stress associated with HCC progression.
Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , RNA, Messenger/genetics , Aged , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Heme Oxygenase-1/genetics , Humans , Liver Neoplasms/blood , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Membrane Glycoproteins/genetics , Middle Aged , NADPH Oxidase 2 , NADPH Oxidases/genetics , Oxidative Stress/genetics , Oxidoreductases Acting on CH-CH Group Donors/blood , Signal Transduction , Up-RegulationABSTRACT
Cyclooxygenase-2 (COX-2) influences carcinogenesis through regulation of angiogenesis, apoptosis, cytokine expression, and immune response suppression. It has been well established that COX-2 is overexpressed in a variety of human cancers, such as hepatocellular carcinoma (HCC). In this study, we aimed to evaluate the association between COX-2 polymorphisms and prognosis of HCC. We genotyped 200 HCC patients of Chinese Han descent for COX-2 gene polymorphisms (-765G>C and -1195G>A) using PCR-RFLP. Data were statistically analyzed using the Kaplan-Meier method and the Cox's proportional hazard regression model. We found that patients with the COX-2 -1195AG and -1195AG + AA genotypes demonstrated significantly decreased disease-free survival (DFS) as compared with those carrying the -1195GG genotype (P < 0.05). However, the COX-2 -765G>C polymorphism was not associated with DFS (P > 0.05). Moreover, by Cox regression analysis, blood alpha fetoprotein ≤400 ng/mL before the operation and the -1195G>A polymorphism were found to be of prognostic significance (P < 0.05), while the -765G>C polymorphism was not (P > 0.05). In summary, post-operation progression of HCC is more likely to occur in patients with the -1195AG genotype and the A allele. On the other hand, the -765G>C polymorphism is not an independent influence factor of HCC prognosis.
Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Matrix Metalloproteinase 2/genetics , Adult , Aged , Alleles , Asian People/genetics , Carcinoma, Hepatocellular/enzymology , China , Cohort Studies , Female , Genetic Predisposition to Disease , Humans , Liver Neoplasms/enzymology , Male , Middle Aged , Polymorphism, Single Nucleotide , PrognosisABSTRACT
Liver cancer is a common malignant tumor associated with a short-survival period and high-mortality rate, and its prevalence in China is particularly high. This study aimed to investigate the effect of overexpressing the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) gene on liver cancer cell apoptosis and provide new insight into the treatment of this disease. The experimental design included four treatment groups, consisting of HHCC and H22 cells transfected with PTEN recombinant plasmids (HHCC+PTEN, H22+PTEN), and those transfected with control plasmids (HHCC+NC, H22+NC). The expression of PTEN mRNA was determined by quantitative polymerase chain reaction, and protein levels were examined by western blot. Cell apoptosis was measured using flow cytometry and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling. PTEN mRNA expression in cells transfected with pcDNA3.1-PTEN was significantly increased compared to the control groups (P < 0.05). In addition, western blotting revealed PTEN protein expression in the treatment groups to be significantly elevated in comparison to control cells (P < 0.05). Flow cytometry showed that apoptosis rates of both HHCC+PTEN (approximately 21.9%) and H22+PTEN (approximately 41.0%) cells were significantly higher than those of the control groups (P < 0.05). Moreover, the difference in apoptosis rate between experimental and control groups was significant (P < 0.05). In this study, HHCC and H22 cells were successfully transfected with pcDNA3.1-PTEN in vitro. We conclude that overexpression of PTEN can effectively inhibit proliferation of these cells and promote their apoptosis.
Subject(s)
Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , PTEN Phosphohydrolase/biosynthesis , Animals , Apoptosis/physiology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , China , Humans , In Situ Nick-End Labeling , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , PTEN Phosphohydrolase/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection/methodsABSTRACT
The signaling pathways that govern survival response in hepatic cancer cells subjected to nutritional restriction have not been clarified yet. In this study we showed that liver cancer cells undergoing glucose deprivation both arrested in G0/G1 and died mainly by apoptosis. Treatment with the AMPK activator AICAR phenocopied the effect of glucose deprivation on cell survival, whereas AMPK silencing in HepG2/C3A, HuH-7 or SK-Hep-1 cells blocked the cell cycle arrest and the increase in apoptotic death induced by glucose starvation. Both AMPK and PKA were promptly activated after glucose withdrawal. PKA signaling had a dual role during glucose starvation: whereas it elicited an early decreased in cell viability, it later improved this parameter. We detected AMPK phosphorylation (AMPKα(Ser173)) by PKA, which was increased in glucose starved cells and was associated with diminution of AMPK activation. To better explore this inhibitory effect, we constructed a hepatocarcinoma derived cell line which stably expressed an AMPK mutant lacking that PKA phosphorylation site: AMPKα1(S173C). Expression of this mutant significantly decreased viability in cells undergoing glucose starvation. Furthermore, after 36 h of glucose deprivation, the index of AMPKα1(S173C) apoptotic cells doubled the apoptotic index observed in control cells. Two main remarks arise: 1. AMPK is the central signaling kinase in the scenario of cell cycle arrest and death induced by glucose starvation in hepatic cancer cells; 2. PKA phosphorylation of Ser173 comes out as a strong control point that limits the antitumor effects of AMPK in this situation.
Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose/deficiency , Liver Neoplasms/metabolism , Apoptosis/physiology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Glucose/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Signal TransductionABSTRACT
BACKGROUND: Ubiquitin Specific Peptidase 39 (USP39) is a 65 kDa SR-related protein involved in RNA splicing. Previous studies showed that USP39 is related with tumorigenesis of human breast cancer cells. RESULTS: In the present study, we investigated the functions of USP39 in human hepatocellular carcinoma (HCC) cell line SMMC-7721. We knocked down the expression of USP39 through lentivirus mediated RNA interference. The results of qRT-PCR and western blotting assay showed that both the mRNA and protein levels were suppressed efficiently after USP39 specific shRNA was delivered into SMMC-7721 cells. Cell growth was significantly inhibited as determined by MTT assay. Crystal violet staining indicated that colony numbers and sizes were both reduced after knock-down of USP39. Furthermore, suppression of USP39 arrested cell cycle progression at G2/M phase in SMMC-7721cells. In addition, Annexin V showed that downregulation of USP39 significantly increased the population of apoptotic cells. CONCLUSIONS: All our results suggest that USP39 is important for HCC cell proliferation and is a potential target for molecular therapy of HCC.
Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Proliferation , Lentivirus/genetics , Liver Neoplasms/pathology , Neoplasm Proteins/metabolism , RNA Interference/physiology , Ubiquitin-Specific Proteases/metabolism , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/enzymology , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Gene Silencing , Gene Transfer Techniques , Humans , In Vitro Techniques , Liver Neoplasms/enzymology , Neoplasm Proteins/genetics , Real-Time Polymerase Chain Reaction , Ubiquitin-Specific Proteases/geneticsABSTRACT
Acetylcholinesterase (AChE), the enzyme that rapidly splits acetylcholine into acetate and choline, presents non-cholinergic functions through which may participate in the control of cell proliferation and apoptosis. These two features are relevant in cancer, particularly in hepatocellular carcinoma (HCC), a very aggressive liver tumor with high incidence and poor prognosis in advanced stages. Here we explored the relation between acetylcholinesterase and HCC growth by testing the influence of AChE on proliferation of Huh-7 and HepG2 cell lines, addressed in monolayer cultures, spheroid formation and human liver tumor samples. Results showed a clear relation in AChE expression and cell cycle progression, an effect which depended on cell confluence. Inhibition of AChE activity led to an increase in cell proliferation, which was associated with downregulation of p27 and cyclins. The fact that Huh-7 and HepG2 cell lines provided similar results lent weight to the relationship of AChE expression with cell cycle progression in hepatoma cell lines at least. Human liver tumor samples exhibited a decrease in AChE activity as compared with normal tissue. The evidence presented herein provides additional support for the proposed tumor suppressor role of AChE, which makes it a potential therapeutic target in therapies against hepatocellular carcinoma.
Subject(s)
Acetylcholinesterase/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Liver Neoplasms/metabolism , Acetylcholinesterase/genetics , Carcinoma, Hepatocellular/enzymology , Cyclins/genetics , Cyclins/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolismABSTRACT
BACKGROUND: Though there is considerable evidence that sphingosine kinase 1(SPHK1) plays a key role in hepatocellular carcinoma(HCC) progression, the prognostic value of SPHK1 expression in HCC with portal vein tumor thrombus (PVTT) remains unclear. Aims. The purpose of this study was to investigate the relationship of SPHK1 expression with PVTT and HCC recurrence after hepatectomy. METHODS: After screening of gene expression profiling of tumor cell lines, real-time PCR and immunohistochemistry were used to investigate the SPHK1 expression in PVTT and HCC samples. The clinical data of 199 HCC patients with nonmain PVTT who underwent liver resection with curative intention were studied. RESULTS: We identified SPHK1 as the most over-expressed gene in PVTT via gene expression profiling of one human PVTT cell line (CSQT-2). SPHK1 expression was an independent factor affecting survival (hazard ratio [HR] 1.799, 95% confidence interval [CI] 1.337-2.368, P < 0.001) and tumor recurrence (HR 1.451, 95% CI 1.087-1.935, P = 0.011). Patients with SPHK1 over-expression had a poorer prognosis than those with SPHK1 under-expression (P < 0.001 and P = 0.011 for survival and tumor recurrence). CONCLUSIONS: SPHK1 might represent a novel and useful prognostic marker of HCC progression in patients with PVTT.
Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Portal Vein , Venous Thrombosis/enzymology , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Disease Progression , Disease-Free Survival , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hepatectomy , Humans , Kaplan-Meier Estimate , Liver Neoplasms/complications , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/surgery , Male , Middle Aged , Neoplasm Recurrence, Local , Phosphotransferases (Alcohol Group Acceptor)/genetics , Portal Vein/pathology , Portal Vein/surgery , Proportional Hazards Models , Prospective Studies , Signal Transduction , Time Factors , Treatment Outcome , Up-Regulation , Venous Thrombosis/etiology , Venous Thrombosis/genetics , Venous Thrombosis/mortality , Venous Thrombosis/surgeryABSTRACT
BACKGROUND: Ubiquitin Specific Peptidase 39 (USP39) is a 65 kDa SR-related protein involved in RNA splicing. Previous studies showed that USP39 is related with tumorigenesis of human breast cancer cells. RESULTS: In the present study, we investigated the functions of USP39 in human hepatocellular carcinoma (HCC) cell line SMMC-7721. We knocked down the expression of USP39 through lentivirus mediated RNA interference. The results of qRT-PCR and western blotting assay showed that both the mRNA and protein levels were suppressed efficiently after USP39 specific shRNA was delivered into SMMC-7721 cells. Cell growth was significantly inhibited as determined by MTT assay. Crystal violet staining indicated that colony numbers and sizes were both reduced after knock-down of USP39. Furthermore, suppression of USP39 arrested cell cycle progression at G2/M phase in SMMC-7721cells. In addition, Annexin V showed that downregulation of USP39 significantly increased the population of apoptotic cells. CONCLUSIONS: All our results suggest that USP39 is important for HCC cell proliferation and is a potential target for molecular therapy of HCC.
Subject(s)
Humans , Cell Cycle , Carcinoma, Hepatocellular/pathology , Lentivirus/genetics , RNA Interference/physiology , Cell Proliferation , Ubiquitin-Specific Proteases/metabolism , Liver Neoplasms/pathology , Neoplasm Proteins/metabolism , In Vitro Techniques , Gene Expression Regulation, Neoplastic/genetics , Cell Cycle/genetics , Blotting, Western , Apoptosis , Gene Transfer Techniques , Carcinoma, Hepatocellular/enzymology , Gene Silencing , Cell Line, Tumor , Cell Proliferation/genetics , Gene Knockdown Techniques , Real-Time Polymerase Chain Reaction , Ubiquitin-Specific Proteases/genetics , Liver Neoplasms/enzymology , Neoplasm Proteins/geneticsABSTRACT
Introduction: Contrast induced nephropathy (CIN) is one of the complications of the use of intravascular contrast agents, being defined as a reduction of the glomerular filtration rate caused by the iodinated contrast. Most CIN data derive from the cardiovascular literature, which identified as the most consistent risk factors pre-existing chronic renal insufficiency and diabetes mellitus. However, these studies limit their conclusions to a more specific patient population. Computerized tomography as a cause of CIN has been studied less often. Objective: To report on the incidence of computerized tomography contrast induced nephropathy (CIN) in an inpatient population of a tertiary general hospital, identifying potentially avoidable risk factors. Methods: We performed a prospective cohort study with inpatients admitted at a tertiary hospital requiring contrast-induced CT. The primary outcome was the development of CIN, measure by the alteration of serum creatinine or glomerular filtration rate in 48 or 72 hours. Through clinical interview, we verified possible risk factors and preventive measures instituted by the medical team and their association with development of CIN. Results: Of a total of 410 patients, 35 (8.5%) developed CIN. There was a positive correlation between CIN and the presence of diabetes mellitus (OR = 2.15; 95%CI 1.35-4.06; p = 0.02), heart failure (OR = 2.23; 95%CI 1.18-8.8; p = 0.022), and renal failure (OR = 3.36; 95%CI 1.57- 7.17; p = 0.002) Conclusion: Incidence of CIN varies according to the population. Diabetes mellitus, heart failure and renal failure were independent risk factors for the development of CT-associated CIN. Further studies are needed to better understand and treat CT-associated CIN. .
Introdução: Nefropatia induzida por contraste (NIC) é consequência do uso de meios de contraste intravenoso, sendo definida como uma redução da taxa de filtração glomerular. A maioria dos dados de NIC são da literatura cardiovascular, que identificou como fatores de risco insuficiência renal crônica e diabetes. Entretanto, esses estudos limitam suas conclusões a uma população especifica de pacientes. Tomografia Computadorizada contrastada como causa de NIC foi menos estudada. Objetivo: Reportar incidência de NIC numa população de pacientes internados em hospital terciário submetidos à tomografia computadorizada com contraste, identificando possíveis fatores de risco evitáveis. Métodos: Realizamos um estudo de coorte prospectivo com pacientes internados em hospital terciário e que necessitaram de tomografia computadorizada com contraste. O desfecho primário foi desenvolvimento de NIC, verificado por meio da variação da creatinina sérica ou taxa de filtração glomerular em 48 ou 72 horas. Em entrevista clínica, verificamos possíveis fatores de risco, assim como medidas preventivas instituídas pela equipe médica e suas possíveis associações com desenvolvimento de NIC. Resultados: Do total de 410 pacientes, 35 (8,5%) desenvolveram NIC. Houve correlação positiva entre desenvolvimento de NIC e a presença de diabetes mellitus (OR = 2,15; 95%CI 1,35-4,06; p = 0,02), insuficiência cardíaca (OR = 2,23; 95%CI 1,18-8,8; p = 0,022), e insuficiência renal (OR = 3,36; 95%CI 1,57-7,17; p = 0,002). Conclusão: A incidência de NIC varia de acordo com a população. Diabetes, insuficiência cardíaca e insuficiência renal foram fatores de risco independentes para o desenvolvimento de NIC. Mais estudos são ...