Gene expression profiling of Human Hepatocellular Carcinoma tissues
Ontology highlight
ABSTRACT: Hepatocellular carcinoma (HCC) is the fifth most common malignancy of cancer-related deaths worldwide, and prognosis of patients with HCC still remain unsatisfactory. Therefore, more reliable biomarkers for predicting the disease and understanding the mechanisms underlying cancer development need to be found out urgently. In this study, we explored the clinical significance and role of ALDOB, G6PD in HCC pathogenesis. Here we show that low ALDOB expression with high G6PD expression lead to patients' poor survival and prognosis by remodeling metabolic pathway and increasing tumor proliferation and metastasis. Stat5a regulates ALDOB expression positively but negatively to G6PD expression in mRNA level, simultaneously, ALDOB inhibits G6PD enzymatic activity by protein-protein interaction and we have found one of their interaction sites in ALDOB. These data suggest that ALDOB and G6PD could be potential biomarkers for HCC.
Project description:Hepatocellular carcinoma (HCC) is the fifth most common malignancy of cancer-related deaths worldwide, and prognosis of patients with HCC still remain unsatisfactory. Therefore, more reliable biomarkers for predicting the disease and understanding the mechanisms underlying cancer development need to be found out urgently. In this study, we explored the clinical significance and role of ALDOB, G6PD in HCC pathogenesis. Here we show that low ALDOB expression with high G6PD expression lead to patients' poor survival and prognosis by remodeling metabolic pathway and increasing tumor proliferation and metastasis. Stat5a regulates ALDOB expression positively but negatively to G6PD expression in mRNA level, simultaneously, ALDOB inhibits G6PD enzymatic activity by protein-protein interaction and we have found one of their interaction sites in ALDOB. These data suggest that ALDOB and G6PD could be potential biomarkers for HCC. We analyzed tissues from 5 hepatocellular carcinoma patients using the GeneChip PrimeView Human Gene Expression Array. Affymetrix microarray assays were performed according to the manufacturer's directions on total RNA isolated from patients' tissues. Array data was processed by Affymetrix Exon Array Computational Tool. No techinical replicates were performed.
Project description:In order to analyze the changes of G6PD enzyme activity regulating the metabolism and proliferation pathways in HCC cell. After G6PD enzyme activity inhibitor RRx-1 treated with 20μM concentrations for 24h in Huh7 cell. Differential expression of genes were selected after RNA sequencing. Then David Bioinformatics was utilized to identify the most significantly regulated GOTERMs and KEGG pathways based on the transcriptional changes.
Project description:Oxaliplatin-based therapeutics is a widely used treatment approach for hepatocellular carcinoma (HCC) patients; however, drug resistance poses a significant clinical challenge. Epigenetic modifications have been implicated in the development of drug resistance. In our study, employing siRNA library screening, we identified that silencing the m6A writer METTL3 significantly enhanced the sensitivity to oxaliplatin in both in vivo and in vitro HCC models. Further investigations through combined RNA-seq and non-targeted metabolomics analysis revealed that silencing METTL3 impeded the pentose phosphate pathway (PPP), leading to a reduction in NADPH and nucleotide precursors. This disruption induced DNA damage, decreased DNA synthesis, and ultimately resulted in cell cycle arrest. Mechanistically, METTL3 was found to modify E3 ligase TRIM21 near the 3'UTR with N6-methyladenosine, leading to reduced RNA stability upon recognition by YTHDF2. TRIM21, in turn, facilitated the degradation of the rate-limiting enzyme of PPP, G6PD, through the ubiquitination-proteasome pathway. Importantly, high expression of METTL3 was significantly associated with adverse prognosis and oxaliplatin resistance in HCC patients. Notably, treatment with the specific METTL3 inhibitor, STM2457, significantly improved the efficacy of oxaliplatin. These findings underscore the critical role of the METTL3/TRIM21/G6PD axis in driving oxaliplatin resistance and present a promising strategy to overcome chemoresistance in HCC.
Project description:Hepatocellular carcinoma (HCC) is a leading cause of death from cancer malignancies. Recently, a putative fifth hexokinase, hexokinase domain containing 1 (HKDC1), was shown to have significant overexpression in HCC compared to healthy tissue. Using a combination of in vitro and in vivo tools, we examined the role of HKDC1 in HCC development and progression. Importantly, HKDC1 ablation stops HCC development and progression via action by promoting metabolic reprogramming and a shift of glucose flux away from the TCA cycle. HKDC1 ablation leads to mitochondrial dysfunction resulting in less cellular energy which cannot be compensated by enhanced glucose uptake. Moreover, we show that the interaction of HKDC1 with the mitochondria is essential for its role in HCC progression, and without this mitochondrial interaction mitochondrial dysfunction occurs. HKDC1 is highly expressed in HCC cells, but only to a minimal degree in hepatocytes under normal conditions therefore targeting HKDC1, specifically its interaction with the mitochondria, reveals a highly selective approach to target cancer cells in HCC.
Project description:Hepatocellular carcinoma (HCC) is a complex and deadly disease lacking druggable genetic mutations. The limited efficacy of systemic treatments for advanced HCC implies that novel predictive biomarkers and drug targets are urgently needed. Most HCC drugs target protein kinases and their kinase-dependent signaling networks to drive HCC progression. To identify HCC signaling networks that determine responses to kinase inhibitors (KIs), we apply a pharmacoproteomics approach integrating kinome activity in 17 HCC cell lines with their responses to 299 KIs, resulting in a comprehensive dataset of pathway-based drug response signatures. By profiling patient HCC samples, we identify signatures of clinical HCC drug responses in individual tumors. Our analyses reveal kinase networks promoting the epithelial-mesenchymal transition (EMT) and drug resistance, including a FZD2-AXL-NUAK1/2 signaling module, whose inhibition reverses the EMT and sensitizes HCC cells to drugs. Our approach identifies cancer drug targets and molecular signatures of drug response for personalized oncology.
Project description:We studied 20 candidate genes (HCC hub genes, potential drug target genes, predominant somatic mutant genes) retrieved from available literature and public databases with potential to be used as biomarkers for the prognosis of HCC and prediction of treatment outcome. We analysed expression of the genes by RT-qPCR in 30 HCC tumor and adjacent non-tumor paired samples from Vietnamese patients.
Project description:Gene-expression profiles of hepatitis C-related, early-stage liver cirrhosis Background & Aims: Liver cirrhosis affects 1%M-bM-^HM-^R2% of population and is the major risk factor of hepatocellular carcinoma (HCC). Hepatitis C cirrhosis-related HCC is the most rapidly increasing cause of cancer death in the US. Non-invasive methods have been developed to identify patients with asymptomatic, early-stage cirrhosis, increasing the burden of HCC surveillance, but biomarkers are needed to identify patients with cirrhosis who are most in need of surveillance. We investigated whether a liver-derived 186-gene signature previously associated with outcomes of patients with HCC is prognostic for patients newly diagnosed with cirrhosis but without HCC. Methods: We performed gene expression profile analysis of formalin-fixed needle biopsies from the livers of 216 patients with hepatitis C-related early-stage (Child-Pugh class A) cirrhosis who were prospectively followed for a median of 10 years at an Italian center. We evaluated whether the 186-gene signature was associated with death, progression of cirrhosis, and development of HCC. Results: Fifty-five (25%), 101 (47%), and 60 (28%) patients were classified as having poor-, intermediate-, and good-prognosis signatures, respectively. In multivariable Cox regression modeling, the poor-prognosis signature was significantly associated with death (P=.004), progression to advanced cirrhosis (P<.001), and development of HCC (P=.009). The 10-year rates of survival were 63%, 74%, and 85% and the annual incidences of HCC were 5.8%, 2.2%, and 1.5% for patients with poor-, intermediate-, and good-prognosis signatures, respectively. Conclusions: A 186-gene signature used to predict outcomes of patients with HCC is also associated with outcomes of patients with hepatitis C-related early-stage cirrhosis. This signature might be used to identify patients with cirrhosis in most need of surveillance and strategies to prevent their development of HCC. 216 liver biopsy specimens
Project description:Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide, indicating urgent need for preventive strategies. Cancer chemoprevention discovery is challenged by the absence of tractable and clinically relevant human model systems and the complex cell circuits supporting carcinogenesis. Here, we developed a simple and robust human liver cell-based system in which persistent hepatitis B/C virus infection or ethanol induce an HCC risk signature robustly predicting long-term HCC risk in cirrhotic patients. Using a computationally enriched small molecule cell-based screen followed by ex/in vivo validation in human liver tissues and animal models, we identified captopril as an HCC chemopreventive agent that antagonizes the EGF/EGFR pathway, a key driver of liver disease and hepatocarcinogenesis. Our system, modeling the cell circuits encoded in the clinical HCC risk signature, enables fast-track cancer chemoprevention discovery and provides an urgently needed approach for improving the dismal prognosis of patients at risk of HCC.
Project description:Hepatocellular carcinoma (HCC) is not only the fifth most prevalent cancer, presenting a major global health problem, but also among the leading causes of cancer-related deaths worldwide as its therapeutic targets are limited. To identify novel therapeutic targets, elucidate its oncogenic activities and molecular mechanism in HCC is urgent. We used R language edgeR package screened the expression profiles of 374 tissue samples obtained from patients with HCC and 50 samples of normal liver tissues obtained from The Cancer Genome Atlas (TCGA) database. Focusing on DDX24, we explored the functional effect and clinical significance of DDX24 in HCC. We provided evidence that DDX24 was a potential pro-tumorigenic gene in HCC. DDX24 knockdown inhibited HCC cell growth in vitro and in vivo. Mechanistically, RFX8 was proved to be DDX24 promoter-binding protein that transcriptionally upregulated DDX24 expression. Furthermore, we found that DDX24 bound to, and increased the stability of, LAMB1 mRNA by RNA immunoprecipitation (RIP) sequencing and RNA sequencing. Survival analysis indicated that HCC patients with high DDX24, RFX8, or LAMB1 expression exhibited poor prognosis. Our results demonstrated that DDX24 promoted HCC via RFX8/DDX24/LAMB1 pathway, which can be exploited as potential therapeutic target against HCC.
Project description:Hepatocellular carcinoma (HCC) is not only the fifth most prevalent cancer, presenting a major global health problem, but also among the leading causes of cancer-related deaths worldwide as its therapeutic targets are limited. To identify novel therapeutic targets, elucidate its oncogenic activities and molecular mechanism in HCC is urgent. We used R language edgeR package screened the expression profiles of 374 tissue samples obtained from patients with HCC and 50 samples of normal liver tissues obtained from The Cancer Genome Atlas (TCGA) database. Focusing on DDX24, we explored the functional effect and clinical significance of DDX24 in HCC. We provided evidence that DDX24 was a potential pro-tumorigenic gene in HCC. DDX24 knockdown inhibited HCC cell growth in vitro and in vivo. Mechanistically, RFX8 was proved to be DDX24 promoter-binding protein that transcriptionally upregulated DDX24 expression. Furthermore, we found that DDX24 bound to, and increased the stability of, LAMB1 mRNA by RNA immunoprecipitation (RIP) sequencing and RNA sequencing. Survival analysis indicated that HCC patients with high DDX24, RFX8, or LAMB1 expression exhibited poor prognosis. Our results demonstrated that DDX24 promoted HCC via RFX8/DDX24/LAMB1 pathway, which can be exploited as potential therapeutic target against HCC.