Journal articles on the topic 'Highly Upregulated in Liver Carcinoma'

To see the other types of publications on this topic, follow the link: Highly Upregulated in Liver Carcinoma.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Highly Upregulated in Liver Carcinoma.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Wadkin, James C. R., Daniel A. Patten, Sivesh K. Kamarajah, Emma L. Shepherd, Vera Novitskaya, Fedor Berditchevski, David H. Adams, Chris J. Weston, and Shishir Shetty. "CD151 supports VCAM-1-mediated lymphocyte adhesion to liver endothelium and is upregulated in chronic liver disease and hepatocellular carcinoma." American Journal of Physiology-Gastrointestinal and Liver Physiology 313, no. 2 (August 1, 2017): G138—G149. http://dx.doi.org/10.1152/ajpgi.00411.2016.

Full text
Abstract:
CD151, a member of the tetraspanin family of receptors, is a lateral organizer and modulator of activity of several families of transmembrane proteins. It has been implicated in the development and progression of several cancers, but its role in chronic inflammatory disease is less well understood. Here we show that CD151 is upregulated by distinct microenvironmental signals in a range of chronic inflammatory liver diseases and in primary liver cancer, in which it supports lymphocyte recruitment. CD151 was highly expressed in endothelial cells of the hepatic sinusoids and neovessels developing in fibrotic septa and tumor margins. Primary cultures of human hepatic sinusoidal endothelial cells (HSECs) expressed CD151 at the cell membrane and in intracellular vesicles. CD151 was upregulated by VEGF and HepG2 conditioned media but not by proinflammatory cytokines. Confocal microscopy confirmed that CD151 colocalized with the endothelial adhesion molecule/immunoglobulin superfamily member, VCAM-1. Functional flow-based adhesion assays with primary human lymphocytes and HSECs demonstrated a 40% reduction of lymphocyte adhesion with CD151 blockade. Inhibition of lymphocyte adhesion was similar between VCAM-1 blockade and a combination of CD151/VCAM-1 blockade, suggesting a collaborative role between the two receptors. These studies demonstrate that CD151 is upregulated within the liver during chronic inflammation, where it supports lymphocyte recruitment via liver endothelium. We propose that CD151 regulates the activity of VCAM-1 during lymphocyte recruitment to the human liver and could be a novel anti-inflammatory target in chronic liver disease and hepatocellular cancer prevention. NEW & NOTEWORTHY Chronic hepatitis is characterized by lymphocyte accumulation in liver tissue, which drives fibrosis and carcinogenesis. Here, we demonstrate for the first time that the tetraspanin CD151 supports lymphocyte adhesion to liver endothelium. We show that CD151 is upregulated in chronic liver disease and hepatocellular carcinoma (HCC) and is regulated on endothelium by tissue remodeling and procarcinogenic factors. These regulatory and functional studies identify CD151 as a potential therapeutic target to treat liver fibrosis and HCC.
APA, Harvard, Vancouver, ISO, and other styles
2

Jiang, Xue, and Weiwei Liu. "Long Noncoding RNA Highly Upregulated in Liver Cancer Activates p53-p21 Pathway and Promotes Nasopharyngeal Carcinoma Cell Growth." DNA and Cell Biology 36, no. 7 (July 2017): 596–602. http://dx.doi.org/10.1089/dna.2017.3686.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Yang, Shuang, Jian Zhou, Weiwu Gao, Xia Yang, Di Yang, Zhiqiang Tian, Yuzhang Wu, Mengjie Zhang, Yi Zhang, and Bing Ni. "Multifunctional YY1 in Liver Diseases." Seminars in Liver Disease 37, no. 04 (November 2017): 363–76. http://dx.doi.org/10.1055/s-0037-1607451.

Full text
Abstract:
AbstractThe transcription factor Yin Yang 1 (YY1) is a multifunctional protein that can activate or repress gene expression, depending on the cellular context. While YY1 is ubiquitously expressed and highly conserved between species, its role varies among the diverse cell types and includes proliferation, differentiation, and apoptosis. Upregulated YY1 expression is found in pathogenic conditions, such as human hepatocellular carcinoma and hepatitis B virus infection, and its roles in the molecular pathogenic mechanisms in liver (i.e., fibrosis, carcinogenesis, viral-induced injury) are currently being elucidated. The most recent studies have revealed that YY1 is deeply involved in such dysregulated cellular metabolisms as glycometabolism, lipid metabolism, and bile acid metabolism, which are all involved in various diseases. In this review, we will summarize the current knowledge on YY1 in liver diseases, providing a focused discussion on the characterized and probable underlying mechanisms, as well as a reasoned evaluation of the potential for YY1-mediated pathology as drug targets in liver disease therapies.
APA, Harvard, Vancouver, ISO, and other styles
4

Henderson, James M., Michelle S. W. Xiang, Jiali Carrie Huang, Stefanie Wetzel, Linxuan Jiang, Jack H. Lai, Wengen Wu, et al. "Dipeptidyl Peptidase Inhibition Enhances CD8 T Cell Recruitment and Activates Intrahepatic Inflammasome in a Murine Model of Hepatocellular Carcinoma." Cancers 13, no. 21 (November 1, 2021): 5495. http://dx.doi.org/10.3390/cancers13215495.

Full text
Abstract:
The mRNA expression of the dipeptidyl peptidase 4 (DPP4) gene family is highly upregulated in human hepatocellular carcinoma (HCC) and is associated with poor survival in HCC patients. Compounds that inhibit the DPP4 enzyme family, such as talabostat and ARI-4175, can mediate tumour regression by immune-mediated mechanisms that are believed to include NLRP1 activation. This study investigated the expression and activity of the DPP4 family during the development of HCC and evaluated the efficacy of ARI-4175 in the treatment of early HCC in mice. This first report on this enzyme family in HCC-bearing mice showed DPP9 upregulation in HCC, whereas intrahepatic DPP8/9 and DPP4 enzyme activity levels decreased with age. We demonstrated that ARI-4175 significantly lowered the total number of macroscopic liver nodules in these mice. In addition, ARI-4175 increased intrahepatic inflammatory cell infiltration, including CD8+ T cell numbers, into the HCC-bearing livers. Furthermore, ARI-4175 activated a critical component of the inflammasome pathway, caspase-1, in these HCC-bearing livers. This is the first evidence of caspase-1 activation by a pan-DPP inhibitor in the liver. Our data suggest that targeting the DPP4 enzyme family may be a novel and effective approach to promote anti-tumour immunity in HCC via caspase-1 activation.
APA, Harvard, Vancouver, ISO, and other styles
5

Ruan, Lingjuan, Lifei Huang, Lilai Zhao, Qiang Wang, Xiaocheng Pan, Anmin Zhang, Qiuping Bai, and Zongjun Lv. "The Interaction of lncRNA-HEIH and lncRNA-HULC with HBXIP in Hepatitis B Patients." Gastroenterology Research and Practice 2018 (December 4, 2018): 1–6. http://dx.doi.org/10.1155/2018/9187316.

Full text
Abstract:
Hepatitis B virus (HBV) infection is a major risk factor for the development of hepatic cirrhosis (HC) and hepatocellular carcinoma (HCC), which are associated with very high morbidity and mortality rates worldwide. Many studies have shown that long noncoding RNAs (lncRNAs) that are highly expressed in HCC (lncRNA-HEIH) and highly upregulated in liver cancer (lncRNA-HULC) have been implicated in the development and progression of hepatitis B-related HC and HCC. In this study, reverse transcription and quantitative PCR were used to detect the expression of lncRNA-HEIH and lncRNA-HULC and western blot analysis to detect the expression of hepatitis B X-interacting protein (HBXIP). RNA immunoprecipitation was used to detect the interaction of HBXIP with lncRNA-HULC and lncRNA-HEIH. The results showed that lncRNA-HEIH, lncRNA-HULC, and HBXIP were upregulated in hepatitis B patients, particularly those with hepatitis B-related HCC. Both lncRNA-HEIH and lncRNA-HULC interacted with HBXIP. These results suggest that lncRNA-HEIH and lncRNA-HULC interact with HBXIP in hepatitis B-related diseases.
APA, Harvard, Vancouver, ISO, and other styles
6

Huynh, Kenneth N., Sriram Rao, Bradley Roth, Theodore Bryan, Dayantha M. Fernando, Farshid Dayyani, David Imagawa, and Nadine Abi-Jaoudeh. "Targeting Hypoxia-Inducible Factor-1α for the Management of Hepatocellular Carcinoma." Cancers 15, no. 10 (May 12, 2023): 2738. http://dx.doi.org/10.3390/cancers15102738.

Full text
Abstract:
Hypoxia-inducible factor 1 alpha (HIF-1α) is a transcription factor that regulates the cellular response to hypoxia and is upregulated in all types of solid tumor, leading to tumor angiogenesis, growth, and resistance to therapy. Hepatocellular carcinoma (HCC) is a highly vascular tumor, as well as a hypoxic tumor, due to the liver being a relatively hypoxic environment compared to other organs. Trans-arterial chemoembolization (TACE) and trans-arterial embolization (TAE) are locoregional therapies that are part of the treatment guidelines for HCC but can also exacerbate hypoxia in tumors, as seen with HIF-1α upregulation post-hepatic embolization. Hypoxia-activated prodrugs (HAPs) are a novel class of anticancer agent that are selectively activated under hypoxic conditions, potentially allowing for the targeted treatment of hypoxic HCC. Early studies targeting hypoxia show promising results; however, further research is needed to understand the effects of HAPs in combination with embolization in the treatment of HCC. This review aims to summarize current knowledge on the role of hypoxia and HIF-1α in HCC, as well as the potential of HAPs and liver-directed embolization.
APA, Harvard, Vancouver, ISO, and other styles
7

Li, Duguang, Junhai Pan, Yiyin Zhang, Yirun Li, Shengxi Jin, Cheng Zhong, Peng Chen, et al. "C8orf76 Modulates Ferroptosis in Liver Cancer via Transcriptionally Up-Regulating SLC7A11." Cancers 14, no. 14 (July 13, 2022): 3410. http://dx.doi.org/10.3390/cancers14143410.

Full text
Abstract:
Hepatocellular carcinoma (HCC) is a common malignant tumor worldwide. Chromosome 8 open reading frame 76 (C8orf76), a novel gene located in the nucleus, is highly expressed in many tumor types. However, the specific mechanisms and functions of C8orf76 in HCC remain unclear. Here, we reported for the first time that C8orf76 gene expression levels were frequently upregulated in liver cancer and significantly correlated with HCC development. C8orf76 downregulation induced G1-S arrest and inhibited cell proliferation. Intriguingly, C8orf76 deficiency could accelerate erastin or sorafenib-induced ferroptosis through increasing lipid reactive oxygen species (ROS) levels. Moreover, although C8orf76 overexpression did not affect tumorigenesis under normal conditions, it increased resistance to lipid disturbance and ferroptosis triggered by erastin or sorafenib, which further facilitated HCC cell growth and tumor progression. Mechanistically, C8orf76 bound to the promoter region of the solute carrier family 7 member 11 (SLC7A11) gene and upregulated SLC7A11 transcriptionally. SLC7A11-dependent cystine import led to sufficient GSH synthesis and lipid peroxidation inhibition, thus accelerating tumor growth. Our study indicated that C8orf76 could be a novel marker for HCC diagnosis. In addition, a better comprehensive understanding of the potential role of C8orf76 in HCC helped us develop novel therapeutic strategies for this intractable cancer.
APA, Harvard, Vancouver, ISO, and other styles
8

Kim, Wantae, Sanjoy Kumar Khan, Yuchen Liu, Ruoshi Xu, Ogyi Park, Yong He, Boksik Cha, Bin Gao, and Yingzi Yang. "Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma." Gut 67, no. 9 (September 2, 2017): 1692–703. http://dx.doi.org/10.1136/gutjnl-2017-314061.

Full text
Abstract:
ObjectiveHippo signalling is a recently identified major oncosuppressive pathway that plays critical roles in inhibiting hepatocyte proliferation, survival and hepatocellular carcinoma (HCC) formation. Hippo kinase (Mst1 and Mst2) inhibits HCC proliferation by suppressing Yap/Taz transcription activities. As human HCC is mainly driven by chronic liver inflammation, it is not clear whether Hippo signalling inhibits HCC by shaping its inflammatory microenvironment.DesignWe have established a genetic HCC model by deleting Mst1 and Mst2 in hepatocytes. Functions of inflammatory responses in this model were characterised by molecular, cellular and FACS analysis, immunohistochemistry and genetic deletion of monocyte chemoattractant protein-1 (Mcp1) or Yap. Human HCC databases and human HCC samples were analysed by immunohistochemistry.ResultsGenetic deletion of Mst1 and Mst2 in hepatocytes (DKO) led to HCC development, highly upregulated Mcp1 expression and massive infiltration of macrophages with mixed M1 and M2 phenotypes. Macrophage ablation or deletion of Mcp1 in DKO mice markedly reduced hepatic inflammation and HCC development. Moreover, Yap removal abolished induction of Mcp1 expression and restored normal liver growth in the Mst1/Mst2 DKO mice. Finally, we showed that MCP1 is a direct transcription target of YAP in hepatocytes and identified a strong gene expression correlation between YAP targets and MCP-1 in human HCCs.ConclusionsHippo signalling in hepatocytes maintains normal liver growth by suppressing macrophage infiltration during protumoural microenvironment formation through the inhibition of Yap-dependent Mcp1 expression, providing new targets and strategies to treat HCCs.
APA, Harvard, Vancouver, ISO, and other styles
9

Correia de Sousa, Marta, Nicolas Calo, Cyril Sobolewski, Monika Gjorgjieva, Sophie Clément, Christine Maeder, Dobrochna Dolicka, et al. "Mir-21 Suppression Promotes Mouse Hepatocarcinogenesis." Cancers 13, no. 19 (October 4, 2021): 4983. http://dx.doi.org/10.3390/cancers13194983.

Full text
Abstract:
The microRNA 21 (miR-21) is upregulated in almost all known human cancers and is considered a highly potent oncogene and potential therapeutic target for cancer treatment. In the liver, miR-21 was reported to promote hepatic steatosis and inflammation, but whether miR-21 also drives hepatocarcinogenesis remains poorly investigated in vivo. Here we show using both carcinogen (Diethylnitrosamine, DEN) or genetically (PTEN deficiency)-induced mouse models of hepatocellular carcinoma (HCC), total or hepatocyte-specific genetic deletion of this microRNA fosters HCC development—contrasting the expected oncogenic role of miR-21. Gene and protein expression analyses of mouse liver tissues further indicate that total or hepatocyte-specific miR-21 deficiency is associated with an increased expression of oncogenes such as Cdc25a, subtle deregulations of the MAPK, HiPPO, and STAT3 signaling pathways, as well as alterations of the inflammatory/immune anti-tumoral responses in the liver. Together, our data show that miR-21 deficiency promotes a pro-tumoral microenvironment, which over time fosters HCC development via pleiotropic and complex mechanisms. These results question the current dogma of miR-21 being a potent oncomiR in the liver and call for cautiousness when considering miR-21 inhibition for therapeutic purposes in HCC.
APA, Harvard, Vancouver, ISO, and other styles
10

Xie, Hui, Hongwei Ma, and Danqiu Zhou. "Plasma HULC as a Promising Novel Biomarker for the Detection of Hepatocellular Carcinoma." BioMed Research International 2013 (2013): 1–5. http://dx.doi.org/10.1155/2013/136106.

Full text
Abstract:
Hepatocellular carcinoma (HCC) is a leading cause of cancer death in many Asian and African countries. Lack of early diagnosis tools is one of the clinical obstacles for effective treatment of HCC. Thus, enhanced understanding of the molecular changes associated with HCC is urgently needed to develop novel strategies for the diagnosis and treatment of this dismal disease. While aberrant expression of long noncoding RNAs (lncRNAs) has been functionally associated with certain cancers, the expression profiles and biological relevance of lncRNAs in HCC remain unclear. Highly upregulated in liver cancer (HULC) lncRNA has been implicated in the regulation of hepatoma cell proliferation. In this study, we demonstrate that HULC expression is significantly higher in HCC tumors compared to normal liver tissues. Among the tumor tissues, higher HULC expression is positively associated with Edmondson histological grades or with hepatitis B (HBV) positive status. Moreover, HULC lncRNA is detected with higher frequency in the plasma of HCC patients compared to healthy controls. Higher HULC detection rates are observed in the plasma of patients with higher Edmondson grades or with HBV+ status. These findings indicate for the first time that the expression of HULC in plasma can be used as a noninvasive promising novel biomarker for the diagnosis and/or prognosis of HCC.
APA, Harvard, Vancouver, ISO, and other styles
11

Liu, Zhenrong, Yaru Wang, Zulihumaer Aizimuaji, Sheng Ma, and Ting Xiao. "Elevated FOXA1 Expression Indicates Poor Prognosis in Liver Cancer due to Its Effects on Cell Proliferation and Metastasis." Disease Markers 2022 (August 5, 2022): 1–16. http://dx.doi.org/10.1155/2022/3317315.

Full text
Abstract:
Purpose. Studying the pathogenesis of liver cancer is conducive to the exploration of effective diagnostic and prognostic biomarkers. In this study, we investigated the expression of FOXA1 and its oncogenic role in hepatocellular carcinoma (HCC). Methods. Transcriptome data of HCC tissues were downloaded from The Cancer Genome Atlas (TCGA) and GEO databases and analyzed using R software. We also upregulated FOXA1 expression in HCC cells and investigated the role of FOXA1 in the proliferation and migration of HCC cells through proliferation, colony formation, wound healing, and Transwell assays. Results. An analysis of the transcriptome data in TCGA database revealed found that FOXA1 is highly expressed in HCC tissues and that patients with low FOXA1 expression have a better prognosis. High FOXA1 expression was mainly associated with extracellular matrix organization, cancer, and mitosis. The results of an immunohistochemistry (IHC) assay showed that FOXA1 protein was highly expressed in HCC tissues, and patients with low FOXA1 expression showed longer disease-specific survival times and progression-free intervals. The results from quantitative reverse transcription–PCR (RT–qPCR) and Western blot experiments showed that the expression of FOXA1 in liver cancer cell lines was higher than that in immortalized human liver cell lines. Proliferation, wound healing, and Transwell experiments showed that FOXA1 enhanced the proliferation and migration abilities of liver cancer and immortalized human cell lines. Conclusion. Our research suggests that FOXA1 plays an important role in promoting the recurrence and metastasis of HCC by increasing cell proliferation and metastasis.
APA, Harvard, Vancouver, ISO, and other styles
12

Han, Sanfeng, Tao Ye, Yuqin Mao, Bo Hu, and Chen Wang. "Cuproptosis-Related Genes CDK1 and COA6 Involved in the Prognosis Prediction of Liver Hepatocellular Carcinoma." Disease Markers 2023 (May 11, 2023): 1–17. http://dx.doi.org/10.1155/2023/5552798.

Full text
Abstract:
Background. Liver hepatocellular carcinoma (LIHC) is the most frequently seen type of primary liver cancer. Cuproptosis is a novel form of cell death highly associated with mitochondrial metabolism. However, the clinical impact and pertinent mechanism of cuproptosis genes in LIHC remain largely unknown. Methods. From public databases, we systematically assessed common genes from LIHC differentially expressed genes (DEGs) and cuproptosis-related genes using bioinformatics analysis. These common genes were then analyzed by enrichment analysis, mutation analysis, risk score model, and others to find candidate hub genes related to LIHC and cuproptosis. Next, hub genes were determined by expression, clinical factors, immunoassay, and prognostic nomogram. Results. Based on 129 cuproptosis-related genes and 3492 LIHC DEGs, we totally identified 21 downregulated and 18 upregulated common genes, and they were enriched in pathways, such as zinc ion homeostasis and oxidative phosphorylation. In the mutation analysis, missense mutation was the most common type in LIHC patients, and the common gene F5 had the highest mutation frequency. After LASSO-Cox regression analysis and prognostic analysis, CDK1, ABCB6, LCAT, and COA6 were identified as prognostic signature genes. Among them, ABCB6 and LCAT were lowly expressed in tumors, and CDK1 and COA6 were highly expressed in tumors. In addition, ABCB6 and LCAT were negatively correlated with 6 kinds of immune cells, while CDK1 and COA6 were positively correlated with them. CDK1 and COA6 were identified as hub genes related to LIHC by Cox regression analysis and prognostic nomogram. Conclusion. CDK1 and COA6 are two oncogenes in LIHC, which are involved in the molecular mechanism of cuproptosis and LIHC. Besides, CDK1 and COA6 can positively regulate the expressions of immune cells in LIHC. In clinical practice, they can be used as immunotherapeutic targets and prognostic predictors in LIHC, which sheds new light on the scientific fields of cuproptosis and LIHC.
APA, Harvard, Vancouver, ISO, and other styles
13

Lopez, Samantha, Adithya Anilkumar, Kyle Doxtater, Sudhir Kotnala, Neeraj Chauhan, Murali Yallapu, Meena Jaggi, Subhash Chauhan, and Manish Tripathi. "Abstract 544: Role of an ankyrin domain protein in hepatocellular carcinoma progression." Cancer Research 82, no. 12_Supplement (June 15, 2022): 544. http://dx.doi.org/10.1158/1538-7445.am2022-544.

Full text
Abstract:
Abstract Background: Liver cancer is the third leading cause of mortality attributed to cancer. Hepatocellular carcinoma (HCC) is the most prevalent form among various histological types accounting for 85% to 90% of primary liver cancers. As per Texas Cancer Registry, the Hispanic population had an incidence of 21.2 per 100,000 in Texas. The Rio Grande Valley, located in South Texas, is an underserved area composed highly of Hispanics. It faces multiple disparities that can attribute to increased risk factors of HCC and, thus, a higher liver cancer incidence and mortality. Due to this, there is a need to identify a new, inexpensive, and faster diagnostic biomarker in liver cancer. We have recently identified an extracellular secreted cancer antigen POTE-2, a member of the ankyrin domain-containing POTE gene family. Our preliminary data indicates high POTE-2 expression in HCC tumors. In this study, we will discuss the role of POTE-2 in HCC progression and its associated regulatory pathways. Methods: The Cancer Genome Atlas (TCGA) database of HCC patients (n=371 tumor; n=50 normal) was analyzed. Liver cancer cell lines procured from ATCC were analyzed for POTE-2 mRNA and protein expression through RT-PCR and western blot, respectively. Lentiviral-based plasmids were used for overexpression and knockdown studies for oncogenic assays. Localization of POTE-2, YAP1, and PDL1 was identified with immunofluorescence. Results: Comprehensive analysis of the TCGA database showed increased POTE-2 expression in tumors and is upregulated in all stages of HCC. Lentiviral transduction led to a change in phenotype in oncogenic assays. Modulation of POTE-2 expression led to changes in kinase activity and regulatory pathways. Immunofluorescence showed nuclear localization of YAP1 and PDL1 in the liver cancer cell lines. Conclusion: These studies will help discover novel mechanisms of POTE-2 protein function, signaling pathways, and its role in liver cancer progression. Citation Format: Samantha Lopez, Adithya Anilkumar, Kyle Doxtater, Sudhir Kotnala, Neeraj Chauhan, Murali Yallapu, Meena Jaggi, Subhash Chauhan, Manish Tripathi. Role of an ankyrin domain protein in hepatocellular carcinoma progression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 544.
APA, Harvard, Vancouver, ISO, and other styles
14

Liu, Kairui, Xiaolin Wu, Xian Zang, Zejian Huang, Zeyu Lin, Wenliang Tan, Xiang Wu, Wenrou Hu, Baoqi Li, and Lei Zhang. "Erratum." Oncology Research Featuring Preclinical and Clinical Cancer Therapeutics 28, no. 5 (December 10, 2020): 559–60. http://dx.doi.org/10.3727/096504020x16032056440102.

Full text
Abstract:
Overexpression of the tumor necrosis factor receptor-associated factor 4 (TRAF4) has been detected in many cancer types and is considered to foster tumor progression. However, the role of TRAF4 in hepatocellular carcinoma (HCC) remains elusive. In this study, we found that TRAF4 was highly expressed in HCC cell lines and HCC tissues compared with normal liver cell lines and adjacent noncancerous tissues. TRAF4 overexpression in HCC tissues was correlated with tumor quantity and vascular invasion. In vitro studies showed that TRAF4 was associated with HCC cell migration and invasion. An in vivo study verified that TRAF4 overexpression facilitated metastasis in nude mice. In addition, overexpressed TRAF4 promoted the phosphorylation of Akt and induced Slug overexpression, leading to downregulated E-cadherin and upregulated vimentin, while silencing TRAF4 moderated the phosphorylation of Akt and repressed the expression of Slug, which resulted in upregulated E-cadherin and downregulated vimentin. These effects were inversed after pretreatment of the PI3K/Akt inhibitor LY294002 or overexpression of constitutively active Akt1. Our study demonstrated that TRAF4 was involved in promoting HCC cell migration and invasion. The process was induced by the EMT through activation of the PI3K/Akt signaling pathway.
APA, Harvard, Vancouver, ISO, and other styles
15

Suzuki-Kemuriyama, Noriko, Akari Abe, Sae Nakane, Megumi Yuki, Katsuhiro Miyajima, and Dai Nakae. "Nonalcoholic steatohepatitis-associated hepatocarcinogenesis in mice fed a modified choline-deficient, methionine-lowered, L-amino acid-defined diet and the role of signal changes." PLOS ONE 18, no. 8 (August 3, 2023): e0287657. http://dx.doi.org/10.1371/journal.pone.0287657.

Full text
Abstract:
Nonalcoholic steatohepatitis (NASH) can progress to cirrhosis and even hepatocellular carcinoma (HCC). The incidence of NASH-associated HCC is increasing, posing a serious public health threat. Unfortunately, the underlying pathological mechanisms, including the possible differences between neoplastic and non-neoplastic lesions, remain largely unknown. Previously, we reported a dietary mouse NASH model with a choline-deficient, methionine-lowered, L-amino-acid-defined, high-fat diet containing shortening without trans fatty acids (CDAA-HF-T[−]), which rapidly induces fibrosis and proliferative lesions in the liver. This study aimed to develop a mouse CDAA-HF-T(−) model capable of assessing NASH-associated hepatocarcinogenesis and identifying key signaling factors involved in its underlying mechanisms. Multiple large masses, histopathologically hepatocellular adenomas and carcinomas, and hemangiosarcomas were detected in the liver samples of mice fed CDAA-HF-T(−) for 52 or 63 weeks, along with highly advanced fibrosis and numerous foamy, phagocytic macrophages in the adjacent nontumoral area. Multiple metastatic nodules were found in the lungs of one of the animals, and lymphoid clusters were found in all CDAA-HF-T(−) group mice. In the Ingenuity Pathways Analysis of RNA expression data, the CDAA-HF-T(−) feeding revealed common signal changes in nontumoral and tumoral liver tissues, including increased IL-8 and RhoGTPases signaling and decreased lipid metabolism. Meanwhile, macrophage inflammatory protein 2 (MIP-2) expression levels were upregulated in nontumoral liver tissue from the end of Week 13 of CDAA-HF-T(−) feeding to the end of Week 63. On the other hand, MIP-2 was expressed on macrophages in non-tumor areas and hepatocytes in tumor areas. Therefore, the CDAA-HF-T(−) mouse model is useful for assessing NASH and NASH-associated hepatocarcinogenesis, and IL-8 signaling plays important roles in NASH-associated carcinogenesis and cirrhosis, but it may also play different roles in nontumoral liver tissue and tumorigenesis.
APA, Harvard, Vancouver, ISO, and other styles
16

Lu, I.-Ta, Shih-Chao Lin, Yi-Chia Chu, Ya Wen, You-Cheng Lin, Wen-Chien Cheng, Jyh-Horng Sheu, and Chi-Chien Lin. "(−)-Agelasidine A Induces Endoplasmic Reticulum Stress-Dependent Apoptosis in Human Hepatocellular Carcinoma." Marine Drugs 20, no. 2 (January 29, 2022): 109. http://dx.doi.org/10.3390/md20020109.

Full text
Abstract:
Liver cancers, such as hepatocellular carcinoma (HCC), are a highly prevalent cause of cancer-related deaths. Current treatments to combat liver cancer are limited. (−)-Agelasidine A, a compound isolated from the methanol extract of Agelasnakamurai, a sesquiterpene guanidine derived from sea sponge, has antibacterial activity. We demonstrated its anticancer capabilities by researching the associated mechanism of (−)-agelasidine A in human liver cancer cells. We found that (−)-agelasidine A significantly reduced viability in Hep3B and HepG2 cells, and we determined that apoptosis was involved in the (−)-agelasidine A-induced Hep3B cell deaths. (−)-Agelasidine A activated caspases 9, 8, and 3, as well as PARP. This effect was reversed by caspase inhibitors, suggesting caspase-mediated apoptosis in the (−)-agelasidine A-treated Hep3B cells. Moreover, the reduced mitochondrial membrane potential (MMP) and the release of cytochrome c indicated that the (−)-agelasidine A-mediated mitochondrial apoptosis was mechanistic. (−)-Agelasidine A also increased apoptosis-associated proteins (DR4, DR5, FAS), which are related to extrinsic pathways. These events were accompanied by an increase in Bim and Bax, proteins that promote apoptosis, and a decrease in the antiapoptotic protein, Bcl-2. Furthermore, our results presented that (−)-agelasidine A treatment bridged the intrinsic and extrinsic apoptotic pathways. Western blot analysis of Hep3B cells treated with (−)-agelasidine A showed that endoplasmic reticulum (ER) stress-related proteins (GRP78, phosphorylated PERK, phosphorylated eIF2α, ATF4, truncated ATF6, and CHOP) were upregulated. Moreover, 4-PBA, an ER stress inhibitor, could also abrogate (−)-agelasidine A-induced cell viability reduction, annexin V+ apoptosis, death receptor (DR4, DR5, FAS) expression, mitochondrial dysfunction, and cytochrome c release. In conclusion, by activating ER stress, (−)-agelasidine A induced the extrinsic and intrinsic apoptotic pathways of human HCC.
APA, Harvard, Vancouver, ISO, and other styles
17

Krizanac, Marinela, Paola Berenice Mass Sanchez, Sarah K. Schröder, Ralf Weiskirchen, and Anastasia Asimakopoulos. "Lipid-Independent Regulation of PLIN5 via IL-6 through the JAK/STAT3 Axis in Hep3B Cells." International Journal of Molecular Sciences 24, no. 8 (April 13, 2023): 7219. http://dx.doi.org/10.3390/ijms24087219.

Full text
Abstract:
Perilipin 5 (PLIN5) is a lipid droplet coat protein that is highly expressed in oxidative tissues such as those of muscles, the heart and the liver. PLIN5 expression is regulated by a family of peroxisome proliferator-activated receptors (PPARs) and modulated by the cellular lipid status. So far, research has focused on the role of PLIN5 in the context of non-alcoholic fatty liver disease (NAFLD) and specifically in lipid droplet formation and lipolysis, where PLIN5 serves as a regulator of lipid metabolism. In addition, there are only limited studies connecting PLIN5 to hepatocellular carcinoma (HCC), where PLIN5 expression is proven to be upregulated in hepatic tissue. Considering that HCC development is highly driven by cytokines present throughout NAFLD development and in the tumor microenvironment, we here explore the possible regulation of PLIN5 by cytokines known to be involved in HCC and NAFLD progression. We demonstrate that PLIN5 expression is strongly induced by interleukin-6 (IL-6) in a dose- and time-dependent manner in Hep3B cells. Moreover, IL-6-dependent PLIN5 upregulation is mediated by the JAK/STAT3 signaling pathway, which can be blocked by transforming growth factor-β (TGF-β) and tumor necrosis factor-α (TNF-α). Furthermore, IL-6-mediated PLIN5 upregulation changes when IL-6 trans-signaling is stimulated through the addition of soluble IL-6R. In sum, this study sheds light on lipid-independent regulation of PLIN5 expression in the liver, making PLIN5 a crucial target for NAFLD-induced HCC.
APA, Harvard, Vancouver, ISO, and other styles
18

Wu, Hao, Zhixin Zhang, Xinyu Han, Sai Zhang, Jinrui Zhang, Pinsheng Han, Youcheng Zhang, Yi Bai, and Yamin Zhang. "Upregulated SSB Is Involved in Hepatocellular Carcinoma Progression and Metastasis through the Epithelial-Mesenchymal Transition, Antiapoptosis, and Altered ROS Level Pathway." Oxidative Medicine and Cellular Longevity 2023 (February 4, 2023): 1–16. http://dx.doi.org/10.1155/2023/5207431.

Full text
Abstract:
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors with high morbidity and mortality. Therefore, finding new diagnostic and therapeutic targets is vital for HCC patients. Recent studies have shown that dysregulation of RNA-binding proteins is often associated with cancer progression. Several studies have reported that the RNA-binding protein SSB can promote cancer occurrence and progression and is linked to tumor epithelial-mesenchymal transition (EMT), which could be a new diagnostic marker and therapeutic target. However, the expression and function of SSB in HCC remain to be elucidated. Therefore, this study is aimed at clarifying the expression and biological function of SSB in HCC through bioinformatics analysis combined with in vitro experiments. We found that SSB is highly expressed in HCC and is associated with the poor prognosis of HCC patients, and it can serve as an independent unfavorable prognostic factor. Knockdown of SSB can inhibit the growth of HCC cells in vitro, increase the level of apoptosis and the expression of pro-apoptosis-related proteins, and decrease the expression of antiapoptotic proteins. Meanwhile, SSB knockdown reduced HCC cell invasiveness, and the expression of EMT-related proteins changed significantly. We also found that the gene SSB was associated with the level of oxidative stress in liver cancer cells, and the level of intracellular reactive oxygen species (ROS) increased after knockdown of SSB. The results of bioinformatics analysis also showed that high expression of SSB may affect the effect of checkpoint blockade (ICB) therapy. In conclusion, we found that SSB is highly expressed in HCC and that upregulated SSB can promote the proliferation and metastasis of HCC through antiapoptotic, altered intracellular oxidative stress level, and EMT pathways, which can serve as a new diagnostic marker and therapeutic target, and patients with high SSB expression may not have obvious ICB therapy effect.
APA, Harvard, Vancouver, ISO, and other styles
19

Gouthamchandra, K., Anuj Kumar, Shivaprasad Shwetha, Anirban Mukherjee, Madhavi Chandra, B. Ravishankar, M. N. Khaja, Provash Chandra Sadhukhan, and Saumitra Das. "Serum proteomics of hepatitis C virus infection reveals retinol-binding protein 4 as a novel regulator." Journal of General Virology 95, no. 8 (August 1, 2014): 1654–67. http://dx.doi.org/10.1099/vir.0.062430-0.

Full text
Abstract:
Persistent infection of hepatitis C virus (HCV) can lead to liver cirrhosis and hepatocellular carcinoma, which are currently diagnosed by invasive liver biopsy. Approximately 15–20 % of cases of chronic liver diseases in India are caused by HCV infection. In North India, genotype 3 is predominant, whereas genotype 1 is predominant in southern parts of India. The aim of this study was to identify differentially regulated serum proteins in HCV-infected Indian patients (genotypes 1 and 3) using a two-dimensional electrophoresis approach. We identified eight differentially expressed proteins by MS. Expression levels of one of the highly upregulated proteins, retinol-binding protein 4 (RBP4), was validated by ELISA and Western blotting in two independent cohorts. We also confirmed our observation in the JFH1 infectious cell culture system. Interestingly, the HCV core protein enhanced RBP4 levels and partial knockdown of RBP4 had a positive impact on HCV replication, suggesting a possible role for this cellular protein in regulating HCV infection. Analysis of RBP4-interacting partners using a bioinformatic approach revealed novel insights into the possible involvement of RBP4 in HCV-induced pathogenesis. Taken together, this study provided information on the proteome profile of the HCV-infected Indian population, and revealed a link between HCV infection, RBP4 and insulin resistance.
APA, Harvard, Vancouver, ISO, and other styles
20

Li, Jiahao, Lansi Chen, Jingjing Pang, Chunxiu Yang, Wen Xie, Guoyan Shen, Hongshan Chen, Xiaoyi Li, Shu-Yuan Xiao, and Yueying Li. "Autophagy-Related Gene WD Repeat Domain 45B Promotes Tumor Proliferation and Migration of Hepatocellular Carcinoma through the Akt/mTOR Signaling Pathway." Diagnostics 13, no. 5 (February 27, 2023): 906. http://dx.doi.org/10.3390/diagnostics13050906.

Full text
Abstract:
Hepatocellular carcinoma (HCC) is a highly aggressive malignant tumor. It has been found that autophagy plays a role both as a tumor promoter and inhibitor in HCC carcinogenesis. However, the mechanism behind is still unveiled. This study aims to explore the functions and mechanism of the key autophagy-related proteins, to shed light on novel clinical diagnoses and treatment targets of HCC. Bioinformation analyses were performed by using data from public databases including TCGA, ICGC, and UCSC Xena. The upregulated autophagy-related gene WDR45B was identified and validated in human liver cell line LO2, human HCC cell line HepG2 and Huh-7. Immunohistochemical assay (IHC) was also performed on formalin-fixed paraffin-embedded (FFPE) tissues of 56 HCC patients from our pathology archives. By using qRT-PCR and Western blots we found that high expression of WDR45B influenced the Akt/mTOR signaling pathway. Autophagy marker LC3- II/LC3-I was downregulated, and p62/SQSTM1 was upregulated after knockdown of WDR45B. The effects of WDR45B knockdown on autophagy and Akt/mTOR signaling pathways can be reversed by the autophagy inducer rapamycin. Moreover, proliferation and migration of HCC can be inhibited after the knockdown of WDR45B through the CCK8 assay, wound-healing assay and Transwell cell migration and invasion assay. Therefore, WDR45B may become a novel biomarker for HCC prognosis assessment and potential target for molecular therapy.
APA, Harvard, Vancouver, ISO, and other styles
21

Önlen Güneri, Cansu, Hamza Malik Okuyan, Gülay Gülbol Duran, and Mehmet Demir. "Investigation of LncRNAs expression in patients with hepatitis B virus." Journal of Clinical Medicine of Kazakhstan 19, no. 6 (December 30, 2022): 27–31. http://dx.doi.org/10.23950/jcmk/12662.

Full text
Abstract:
<b>Aim: </b>Patients infected with the hepatitis B virus (HBV) are at a higher risk of cirrhosis and hepatocellular carcinoma. Despite the recent advancement of antiviral therapy, many patients still cannot respond to existing therapies. Hence, to detect the changes in liver function earlier, non-invasive methods are needed. Long non-coding RNAs (lncRNAs) play important roles in essential biological process as well as human cancer. LncRNAs may be used as biomarkers in human diseases. Thus, in this study, we purposed to analyze the expression levels of lncRNAs (HOX transcript antisense RNA (HOTAIR), maternally expressed 3 (MEG-3), highly upregulated in liver cancer&nbsp;(HULC)) in patients with hepatitis B virus and healthy volunteers.<br /> <b>Methods: </b>We selected three lncRNAs as candidate lncRNAs based on their association with liver disease.&nbsp;Whole blood samples were collected from 40 patients with HBV and 48 healthy volunteers. The expression levels of all the samples were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR). Statistical analysis was implemented using GraphPad Prism software. A p-value lower than 0.05 was statistically meaningful.<br /> <b>Results: </b>The expression levels of HOTAIR and HULC were remarkably upregulated in the plasma of the patients with HBV compared with healthy control (p&lt;0.05). In contrast, no significant difference in MEG-3 expression levels was observed between groups.<br /> <b>Conclusion: </b>Our findings showed that the expression of HOTAIR and HULC in plasma might be new promising&nbsp;diagnostic and/or prognostic biomarkers for HBV.
APA, Harvard, Vancouver, ISO, and other styles
22

Wang, Gang, Xuefeng Li, Hongyan Jia, Lei Zhang, Yang Li, Qiuxue Zhang, and Meng Lu. "Curcumin nanocarrier carrying si-TAR-RNA binding protein (TRBP) restricts development of hepatocellular carcinoma." Materials Express 13, no. 3 (March 1, 2023): 407–13. http://dx.doi.org/10.1166/mex.2023.2368.

Full text
Abstract:
This study investigated the role of curcumin nanocarriers carrying TAR-RNA binding protein (TRBP) in hepatocellular carcinoma (HCC). We conducted a retrospective analysis of 60 HCC tissue samples and 30 normal liver tissues. TRBP level was measured in HCC. Disease-free survival (DFS) and overall survival (OS) were calculated to analyze correlation between TRBP expression and diagnostic sensitivity of HCC. Survival factors were analyzed using cyclo-oxygen-ase (COX) univariate and multivariate survival analysis. The curcumin nanocarriers-loaded siRNA (si)-TRBP were transfected into HCC cells during in vitro experiments to detect HCC cell proliferation and invasion, whilst flow cytometry measured apoptosis rate of cancer cells. TRBP was highly expressed in 60 HCC tissues and positively associated with diagnostic sensitivity of HCC, with lower DFS and OS in HCC patients with higher TRBP expression, and FIGO stage and TRBP were indicated as risk factors for DFS and OS. High TRBP expression coexisted with increased HCC cell proliferation and invasion. Moreover, transfection of curcumin nanocarriers-loaded si-TRBP dramatically decreased cell viability and invasion, downregulated cleaved caspase-3 and caspase-9, while upregulating Bcl-2. Collectively, this study demonstrated TRBP is upregulated in HCC and curcumin nanocarriers carrying si-TRBP, significantly suppressing growth of HCC cells, providing a theoretical basis for treatment of HCC.
APA, Harvard, Vancouver, ISO, and other styles
23

Kitab, Bouchra, and Kyoko Tsukiyama-Kohara. "Regulatory Role of Ribonucleotide Reductase Subunit M2 in Hepatocyte Growth and Pathogenesis of Hepatitis C Virus." International Journal of Molecular Sciences 24, no. 3 (January 30, 2023): 2619. http://dx.doi.org/10.3390/ijms24032619.

Full text
Abstract:
Hepatitis C virus (HCV) frequently causes chronic infection in the human liver, which may progress to advanced hepatic fibrosis, cirrhosis, and hepatocellular carcinoma. HCV primarily infects highly differentiated quiescent hepatocytes and can modulate cell cycle-regulatory genes and proliferation pathways, which ultimately contribute to persistent infection and pathogenesis. On the other hand, several studies have shown differential regulation of HCV RNA and viral protein expression levels, depending on the proliferation state of hepatocytes and the phase of the cell cycle. HCV typically requires factors provided by host cells for efficient and persistent viral replication. Previously, we found that HCV infection upregulates the expression of ribonucleotide reductase subunit M2 (RRM2) in quiescent hepatocytes. RRM2 is a rate-limiting protein that catalyzes de novo synthesis of deoxyribonucleotide triphosphates, and its expression is highly regulated during various phases of the cell cycle. RRM2 functions as a pro-viral factor essential for HCV RNA synthesis, but its functional role in HCV-induced liver diseases remains unknown. Here, we present a comprehensive review of the role of the hepatocyte cell cycle, in correlation with RRM2 expression, in the regulation of HCV replication. We also discuss the potential relevance of this protein in the pathogenesis of HCV, particularly in the development of hepatocellular carcinoma.
APA, Harvard, Vancouver, ISO, and other styles
24

Khan, Walizeb, Washaakh Ahmad, Anwar M. Hashem, Shadi Zakai, Shafiul Haque, Muhammad Faraz Arshad Malik, Steve Harakeh, and Farhan Haq. "Genomic Relevance of FGFR2 on the Prognosis of HCV-Induced Hepatocellular Carcinoma Patients." Journal of Clinical Medicine 11, no. 11 (May 30, 2022): 3093. http://dx.doi.org/10.3390/jcm11113093.

Full text
Abstract:
The Fibroblast Growth Factor Receptors (FGFRs) are known to regulate cancer metabolism in different tumor types, including hepatocellular carcinoma (HCC). Several risk factors are associated with HCC, of which viral infections (Hepatitis B and C) and cirrhosis are prominent. In Pakistan as well as in highly developed countries like the United States, hepatitis C virus HCV infections are most commonly reported in HCC. Here, we aimed to investigate the clinical relevance of FGFR receptors in HCC and their role in HCV-positive HCC cases. 264 HCC samples along with their clinical information and 96 normal liver samples were collected. qPCR was done to estimate the expression of FGFR1, FGFR2, FGFR3 and FGFR4. Three independent HCV-induced HCC cohorts (containing 293 HCC samples) were used for validation. According to in vitro results, FGFR1 was upregulated in HCV+ HCC patients. However, in all three independent cohorts of HCC, significant a down-regulation of FGFR1 was observed. FGFR2 overexpression was observed in the in vitro cohort as well as in three independent HCC cohorts. Interestingly, a strong correlation of FGFR2 expression was observed between cirrhosis and HCV in all four HCC cohorts. Our study suggested that FGFR2 expression can be used to classify HCC patients based on HCV infection. This FGFR2-based classification may lead to new therapeutic strategies against HCV-positive HCC subtypes.
APA, Harvard, Vancouver, ISO, and other styles
25

Tao, Hanchuan, Cheng Wang, Chongmei Lu, Ning Ma, Yifan Zhu, Shihai Xuan, and Xiaojun Zhou. "Comprehensive Analysis on the Specific Role and Function of Mitochondrial Inner Membrane Protein MPV17 in Liver Hepatocellular Carcinoma." Genetics Research 2022 (July 19, 2022): 1–14. http://dx.doi.org/10.1155/2022/7236823.

Full text
Abstract:
Background. Liver hepatocellular carcinoma (LIHC) is the predominant type of liver cancer, and its treatment still faces great challenges presently. Mitochondrial inner membrane protein MPV17 is reported to be involved in multiple biological activities of cancers. Here, we seek to investigate the specific role and functions of MPV17 in LIHC progression. Methods. Firstly, MPV17 expressions in various tumors and corresponding normal samples and LIHC groups with various clinical features were analyzed, respectively. Next, the relationship between MPV17 expression and LIHC survival was analyzed and verified by AUC curves. Besides, differentially expressed genes (DEGs) for LIHC were screened from TCGA and then analyzed by GO and KEGG. Then, MPV17 was analyzed by prognostic model, Cox analysis, predictive nomogram, pathway correlation, and immunoassay. Finally, the functions of MPV17 were determined by CCK-8 and Tranwell assays. Results. In most tumors, MPV17 expression was higher than that in the normal group, and it was related to LIHC clinical features. In the LIHC survival analysis, highly expressed MPV17 was associated with a poor prognosis. Besides, 314 upregulated and 193 downregulated DEGs are mainly involved in the TNF signaling pathway and tyrosine metabolism. Through prognostic model, Cox analysis, and predictive nomogram, MPV17 had the prognostic value for LIHC. Gene-pathway correlation analysis showed that MPV17 had the strongest correlation with the G2M_checkpoint pathway. In an immunoassay, MPV17 had a strong correlation with many immune cells. Functional assays showed that MPV17 reduction in LIHC cells could inhibit cell invasion, migration, and proliferation. Conclusion. MPV17, as a tumor promoter, could be a new biomarker for LIHC diagnosis and prognosis and probably shed new light on the exploration of LIHC therapies.
APA, Harvard, Vancouver, ISO, and other styles
26

Li, Jianguo, Jin Zhou, Shuangshuang Kai, Can Wang, Daijun Wang, and Jiying Jiang. "Network-Based Coexpression Analysis Identifies Functional and Prognostic Long Noncoding RNAs in Hepatocellular Carcinoma." BioMed Research International 2020 (October 6, 2020): 1–11. http://dx.doi.org/10.1155/2020/1371632.

Full text
Abstract:
Hepatocellular carcinoma (HCC) is a primary liver cancer associated with a growing incidence and extremely high mortality. However, the pathogenic mechanism is still not fully understood. In the present study, we identified 1,631 upregulated and 1,515 downregulated genes and found that cell cycle and metabolism-related pathways or biological processes highly dysregulated in HCC. To assess the biological importance of these DEGs, we carried out weighted gene coexpression network analysis (WGCNA) to identify the functional modules potentially involved in HCC pathogenesis or progression. The five modules were detected with Dynamic Tree Cut algorithm, and GO enrichment analysis revealed that these modules exhibited different biological processes or signaling pathways, such as metabolism-related pathways, cell proliferation-related pathways, and molecules in tumor microenvironment. Moreover, we also observed two immune cells, namely, cytotoxic cells and macrophage enriched in modules grey and brown, respectively, while T helper cell-2 (Th2) was enriched in module turquoise. Among the WGCNA network, four hub long noncoding RNAs (lncRNAs) were identified to be associated with HCC prognostic outcomes, suggesting that coexpression network analysis could uncover lncRNAs with functional importance, which may be associated with prognostic outcomes of HCC patients. In summary, this study demonstrated that network-based analysis could identify some functional modules and some hub-lncRNAs, which may be critical for HCC pathogenesis or progression.
APA, Harvard, Vancouver, ISO, and other styles
27

Li, Ru, Hikari Okada, Taro Yamashita, Kouki Nio, Han Chen, Yingyi Li, Tetsuro Shimakami, et al. "FOXM1 Is a Novel Molecular Target of AFP-Positive Hepatocellular Carcinoma Abrogated by Proteasome Inhibition." International Journal of Molecular Sciences 23, no. 15 (July 27, 2022): 8305. http://dx.doi.org/10.3390/ijms23158305.

Full text
Abstract:
Alpha-fetoprotein (AFP) is an oncofetal protein that is elevated in a subset of hepatocellular carcinoma (HCC) with poor prognosis, but the molecular target activated in AFP-positive HCC remains elusive. Here, we demonstrated that the transcription factor forkhead box M1 (FOXM1) is upregulated in AFP-positive HCC. We found that FOXM1 expression was highly elevated in approximately 40% of HCC cases, and FOXM1-high HCC was associated with high serum AFP levels, a high frequency of microscopic portal vein invasion, and poor prognosis. A transcriptome and pathway analysis revealed the activation of the mitotic cell cycle and the inactivation of mature hepatocyte metabolism function in FOXM1-high HCC. The knockdown of FOXM1 reduced AFP expression and induced G2/M cell cycle arrest. We further identified that the proteasome inhibitor carfilzomib attenuated FOXM1 protein expression and suppressed cell proliferation in AFP-positive HCC cells. Carfilzomib in combination with vascular endothelial growth factor receptor 2 (VEGFR2) blockade significantly prolonged survival by suppressing AFP-positive HCC growth in a subcutaneous tumor xenotransplantation model. These data indicated that FOXM1 plays a pivotal role in the proliferation of AFP-positive liver cancer cells. Carfilzomib can effectively inhibit FOXM1 expression to inhibit tumor growth and could be a novel therapeutic option in patients with AFP-positive HCC who receive anti-VEGFR2 antibodies.
APA, Harvard, Vancouver, ISO, and other styles
28

Ong, Jiann Ruey, Oluwaseun Adebayo Bamodu, Nguyen Viet Khang, Yen-Kuang Lin, Chi-Tai Yeh, Wei-Hwa Lee, and Yih-Giun Cherng. "SUMO-Activating Enzyme Subunit 1 (SAE1) Is a Promising Diagnostic Cancer Metabolism Biomarker of Hepatocellular Carcinoma." Cells 10, no. 1 (January 17, 2021): 178. http://dx.doi.org/10.3390/cells10010178.

Full text
Abstract:
Hepatocellular carcinoma (HCC) is one of the most diagnosed malignancies and a leading cause of cancer-related mortality globally. This is exacerbated by its highly aggressive phenotype, and limitation in early diagnosis and effective therapies. The SUMO-activating enzyme subunit 1 (SAE1) is a component of a heterodimeric small ubiquitin-related modifier that plays a vital role in SUMOylation, a post-translational modification involving in cellular events such as regulation of transcription, cell cycle and apoptosis. Reported overexpression of SAE1 in glioma in a stage-dependent manner suggests it has a probable role in cancer initiation and progression. In this study, hypothesizing that SAE1 is implicated in HCC metastatic phenotype and poor prognosis, we analyzed the expression of SAE1 in several cancer databases and to unravel the underlying molecular mechanism of SAE1-associated hepatocarcinogenesis. Here, we demonstrated that SAE1 is over-expressed in HCC samples compared to normal liver tissue, and this observed SAE1 overexpression is stage and grade-dependent and associated with poor survival. The receiver operating characteristic analysis of SAE1 in TCGA−LIHC patients (n = 421) showed an AUC of 0.925, indicating an excellent diagnostic value of SAE1 in HCC. Our protein-protein interaction analysis for SAE1 showed that SAE1 interacted with and activated oncogenes such as PLK1, CCNB1, CDK4 and CDK1, while simultaneously inhibiting tumor suppressors including PDK4, KLF9, FOXO1 and ALDH2. Immunohistochemical staining and clinicopathological correlate analysis of SAE1 in our TMU-SHH HCC cohort (n = 54) further validated the overexpression of SAE1 in cancerous liver tissues compared with ‘normal’ paracancerous tissue, and high SAE1 expression was strongly correlated with metastasis and disease progression. The oncogenic effect of upregulated SAE1 is associated with dysregulated cancer metabolic signaling. In conclusion, the present study demonstrates that SAE1 is a targetable cancer metabolic biomarker with high potential diagnostic and prognostic implications for patients with HCC.
APA, Harvard, Vancouver, ISO, and other styles
29

Jang, Jeong Won, Ji Min Kim, Hye Seon Kim, Jin Seoub Kim, Ji Won Han, Soon Kyu Lee, Heechul Nam, et al. "Diagnostic performance of serum exosomal miRNA-720 in hepatocellular carcinoma." Journal of Liver Cancer 22, no. 1 (March 31, 2022): 30–39. http://dx.doi.org/10.17998/jlc.2022.02.25.

Full text
Abstract:
Background/Aim: Hepatocellular carcinoma (HCC) is associated with poor prognosis, largely due to late detection. Highly accurate biomarkers are urgently needed to detect early-stage HCC. Our study aims to explore the diagnostic performance of serum exosomal microRNA (miR)-720 in HCC.Methods: Exosomal miRNA was measured via quantitative real-time PCR. A correlation analysis of exosomal miR-720 and tumor or clinico-demographic data of patients with HCC was performed. The receiver operating characteristic (ROC) curve was used to assess the diagnostic capacity of serum exosomal miR-720 for HCC, in comparison with α-fetoprotein (AFP) and prothrombin induced by vitamin K absence or antagonist-II (PIVKA-II).Results: MiR-720 was chosen as a potential HCC marker via miR microarray based on significant differential expression between tumor and non-tumor samples. Serum exosomal miR-720 was significantly upregulated in patients with HCC (n=114) versus other liver diseases (control, n=30), with a higher area under the ROC curve (AUC=0.931) than the other markers. Particularly, serum exosomal miR-720 showed superior performance in diagnosing small HCC (< 5 cm; AUC=0.930) compared with AFP (AUC=0.802) or PIVKA-II (AUC=0.718). Exosomal miR-720 levels showed marginal correlation with tumor size. The proportion of elevated miR-720 also increased with intrahepatic tumor stage progression. Unlike AFP or PIVKA-II showing a significant correlation with aminotransferase levels, the exosomal miR-720 level was not affected by aminotransferase levels.Conclusions: Serum exosomal miR-720 is an excellent biomarker for the diagnosis of HCC, with better performance than AFP or PIVKA-II. Its diagnostic utility is maintained even in small HCC and is unaffected by aminotransferase levels.
APA, Harvard, Vancouver, ISO, and other styles
30

Yang, Yi, Ming Yang, Huasheng Pang, Yiwen Qiu, Ting Sun, Tao Wang, Shu Shen, and Wentao Wang. "A Macrophage Differentiation-Mediated Gene: DDX20 as a Molecular Biomarker Encompassing the Tumor Microenvironment, Disease Staging, and Prognoses in Hepatocellular Carcinoma." Oxidative Medicine and Cellular Longevity 2022 (October 5, 2022): 1–22. http://dx.doi.org/10.1155/2022/9971776.

Full text
Abstract:
Background. DDX20 involves the mechanism of cell proliferate, mitogenic Ets transcriptional suppressor (METS), which can arrest the cell cycle of macrophages. However, little is known about DDX20 expression, clinical values, and the relationship with tumor microenvironment in HCC. Methods. We mined the transcriptional, protein expression and survival data of DDX20 in HCC from online databases. The immunological effects of DDX20 were estimated by bioinformatic algorithms. The RNAi and CRISPR screening were used to assess the gene effect of DDX20 for the EGFR gene in liver tumor cell. Results. We found that the DDX20 was highly expressed in HCC. The qRT-PCR result shows a significantly upregulated DDX20 expression in HCC samples from the West China Hospital. The high mRNA expression of DDX20 is associated with a poor survival. DDX20 expression is positively correlated with MDSCs in HCC tissues. Moreover, DDX20 has a high predicted ability for the response to immunotherapy. Furthermore, hsa-mir-324-5p could regulate the macrophage differentiation by interacting with DDX20. Meanwhile, the EGFR gene gets a high dependency score for DDX20. Conclusion. In sum, DDX20 may serve as a prognostic marker for worse clinical outcomes with HCC and potentially enable more precise and personalized immunotherapeutic strategies in the future.
APA, Harvard, Vancouver, ISO, and other styles
31

Paslaru, Liliana, Gabriela Bindea, Anca Nastase, Andrei Sorop, Cristian Zimbru, Vlad Herlea, Doina Hrehoret, et al. "Comparative RNA-Sequencing Analysis Reveals High Complexity and Heterogeneity of Transcriptomic and Immune Profiles in Hepatocellular Carcinoma Tumors of Viral (HBV, HCV) and Non-Viral Etiology." Medicina 58, no. 12 (December 7, 2022): 1803. http://dx.doi.org/10.3390/medicina58121803.

Full text
Abstract:
Background and Objectives: Hepatocellular carcinoma (HCC), the most common type of primary liver cancer, is the leading cause of cancer-related mortality. It arises and progresses against fibrotic or cirrhotic backgrounds mainly due to infection with hepatitis viruses B (HBV) or C (HCV) or non-viral causes that lead to chronic inflammation and genomic changes. A better understanding of molecular and immune mechanisms in HCC subtypes is needed. Materials and Methods: To identify transcriptional changes in primary HCC tumors with or without hepatitis viral etiology, we analyzed the transcriptomes of 24 patients by next-generation sequencing. Results: We identified common and unique differentially expressed genes for each etiological tumor group and analyzed the expression of SLC, ATP binding cassette, cytochrome 450, cancer testis, and heat shock protein genes. Metascape functional enrichment analysis showed mainly upregulated cell-cycle pathways in HBV and HCV and upregulated cell response to stress in non-viral infection. GeneWalk analysis identified regulator, hub, and moonlighting genes and highlighted CCNB1, ACTN2, BRCA1, IGF1, CDK1, AURKA, AURKB, and TOP2A in the HCV group and HSF1, HSPA1A, HSP90AA1, HSPB1, HSPA5, PTK2, and AURKB in the group without viral infection as hub genes. Immune infiltrate analysis showed that T cell, cytotoxic, and natural killer cell markers were significantly more highly expressed in HCV than in non-viral tumors. Genes associated with monocyte activation had the highest expression levels in HBV, while high expression of genes involved in primary adaptive immune response and complement receptor activity characterized tumors without viral infection. Conclusions: Our comprehensive study underlines the high degree of complexity of immune profiles in the analyzed groups, which adds to the heterogeneous HCC genomic landscape. The biomarkers identified in each HCC group might serve as therapeutic targets.
APA, Harvard, Vancouver, ISO, and other styles
32

Pár, Alajos, and Gabriella Pár. "Alkoholos májbetegség: a genetikai-epigenetikai tényezők szerepe és az absztinencia hatása." Orvosi Hetilap 160, no. 14 (April 2019): 524–32. http://dx.doi.org/10.1556/650.2019.31352.

Full text
Abstract:
Abstract: The pathogenesis of alcoholic liver disease depends not only on the toxic effects of alcohol, but also on the complex interaction of host’s and environmental factors. Thus, the genetic pre-disposition, co-morbidities and behavioral factors all play a role in the individual variations in the disease outcomes. On the other hand, the essential part of the therapeutic strategy is the complete withdrawal of the harmful etiological agent. The present paper is devoted to overview the genetics, the environmental factors and the effects of abstinence in alcoholic liver disease. Genetic variants in two enzymes involved in the metabolism of ethanol, alcohol-dehydrogenase ADH1B *2 and aldehyde-dehydrogenase ALDH2 *2 through increasing the blood level of acetaldehyde, may play a “protective” role against alcoholism. The P450 CYP2E1 *5 c2, an inducible microsomal oxidase, upregulated by ethanol and by formation of acetaldehyde and reactive oxygen species, increases liver toxicity. Three novel gene polymorphisms – such as the patatin-like phospholipase domain-containing 3 (PNPLA3 I148M C>G), the transmembrane 6 superfamily member 2 (TM6SF2 E167K), and the membrane-bound O-acyltransferase domain-containing 7 (MB0AT7 rs641738 C>T) – have been proven as risk factors of steatosis, fibrosis and even hepatocellular carcinoma in both alcoholic and non-alcoholic fatty liver disease patients. Alcohol-induced epigenetic effects, reversible but inheritable gene expression alterations – as histon modulations, DNA methylation and micro-RNA-s – are of importance in the pathogenesis as well, and in the future, they may serve as diagnostic markers and therapeutic targets. Women are at greater risk of developing alcoholic cirrhosis, furthermore, malnutrition, obesity, diabetes, smoking, and hepatitis virus infections are also risk factors. Alcoholic liver disease should be regarded as a preventable disease. Several clinical studies revealed that abstinence may result in the regression of steatohepatitis and fibrosis, compensation of cirrhosis, improving disease outcome and increasing survival even in patients with advanced stages. Early diagnosis and multidisciplinary interventions are highly required to achieve long-term abstinence and to prevent alcoholic cirrhosis. Orv Hetil. 2019; 160(14): 524–532.
APA, Harvard, Vancouver, ISO, and other styles
33

Yuan, Hongchao, Yuanjun Lu, Yau-Tuen Chan, Cheng Zhang, Ning Wang, and Yibin Feng. "The Role of Protein SUMOylation in Human Hepatocellular Carcinoma: A Potential Target of New Drug Discovery and Development." Cancers 13, no. 22 (November 14, 2021): 5700. http://dx.doi.org/10.3390/cancers13225700.

Full text
Abstract:
Small ubiquitin-like modifier (SUMO) is a highly conserved post-translational modification protein, mainly found in eukaryotes. They are widely expressed in different tissues, including the liver. As an essential post-translational modification, SUMOylation is involved in many necessary regulations in cells. It plays a vital role in DNA repair, transcription regulation, protein stability and cell cycle progression. Increasing shreds of evidence show that SUMOylation is closely related to Hepatocellular carcinoma (HCC). The high expression of SUMOs in the inflammatory hepatic tissue may lead to the carcinogenesis of HCC. At the same time, SUMOs will upregulate the proliferation and survival of HCC, migration, invasion and metastasis of HCC, tumour microenvironment as well as drug resistance. This study reviewed the role of SUMOylation in liver cancer. In addition, it also discussed natural compounds that modulate SUMO and target SUMO drugs in clinical trials. Considering the critical role of SUMO protein in the occurrence of HCC, the drug regulation of SUMOylation may become a potential target for treatment, prognostic monitoring and adjuvant chemotherapy of HCC.
APA, Harvard, Vancouver, ISO, and other styles
34

Li, Yaqun, Wenhuan Fu, Zikai Geng, Yun Song, Xionggang Yang, Tianye He, Jian Wu, and Bin Wang. "A pan-cancer analysis of the oncogenic role of ribonucleotide reductase subunit M2 in human tumors." PeerJ 10 (November 28, 2022): e14432. http://dx.doi.org/10.7717/peerj.14432.

Full text
Abstract:
Background Recent studies have identified ribonucleotide reductase subunit M2 (RRM2) as a putative promoter of tumors. However, no systematic analysis of its carcinogenicity has been conducted. Methods The potential functions of RRM2 in various tumor types were investigated using data from the Genotype-Tissue Expression (GTEx), the Clinical Proteomic Tumor Analysis Consortium (CPTAC), the Cancer Genome Atlas (TCGA), the Human Protein Atlas (HPA), cBioPortal, GEPIA, String, and Gene Set Enrichment Analysis (GSEA). We analyzed the difference in mRNA and protein expression, pathological stage, survival, mutation, tumor microenvironment (TME), and immune cell infiltration in relation to RRM2. Meanwhile, using TCGA and the Tumor Immune Estimation Resource 2 (TIMER 2), the associations between RRM2 expression, immune infiltration, and immune-related genes were assessed. Additionally, CCK-8, Edu and RT-PCR assays were used to validate that RRM2 acts as an oncogene in liver cancer cells and its association with HBx. A cohort of liver hepatocellular carcinoma (LIHC) patients (n=154) from Huashan Hospital was analyzed for the expression of RRM2 and the association between RRM2 and immune infiltration. Results Using the GTEx and TCGA databases, we discovered that 28 tumors expressed RRM2 at significantly higher levels than the corresponding normal tissues. Increased RRM2 expression may be predictive of a poor overall survival (OS) in patients with seven different cancers. GO, KEGG, and GSEA analyses revealed that the biological process of RRM2 was associated with the regulation of carcinogenic processes and immune pathways in a variety of tumor types. The expression of RRM2 was highly correlated with maker genes involved in immune activation and immunosuppression, immune checkpoints, DNA mismatch repair system (MMR), and the infiltration levels of Tregs and macrophages (TAMs), suggesting that the carcinogenic effect of RRM2 may be achieved by regulating immune related genes. Moreover, as demonstrated by CCK-8 and Edu assays, RRM2 was an oncogene in liver cancer cells. We confirmed for the first time that RRM2 was significantly upregulated by HBx, suggesting that RRM2 may be a key regulator of LIHC induced by HBV. IHC analysis validated the upregulated expression of RRM2 protein and its correlation with immune infiltration makers in a LIHC patient cohort. Conclusion RRM2 may be a valuable molecular biomarker for predicting prognosis and immunotherapeutic efficacy in pan-cancer, particularly in LIHC.
APA, Harvard, Vancouver, ISO, and other styles
35

Huang, Shuai, Xiangkun Wang, Kai Luo, Xudong Zhang, Zhongyuan Liu, and Renfeng Li. "Combined Evaluation of mRNA and Protein Expression, Promoter Methylation, and Immune Infiltration of UBE2I in Pan-Digestive System Tumors." Oxidative Medicine and Cellular Longevity 2022 (September 20, 2022): 1–20. http://dx.doi.org/10.1155/2022/1129062.

Full text
Abstract:
Background. Digestive system tumors (DSTs) have high morbidity and mortality worldwide. This study explored the potential value of ubiquitin-conjugating enzyme E2 I (UBE2I) in pan-digestive system tumors (pan-DSTs). Methods. Differential expression, tumor stages, and survival outcomes of UBE2I in pan-DSTs were determined using the GEPIA database. The TIMER database was used to confirm the correlation of UBE2I expression with pan-DSTs and immune infiltrates. Differential analyses of UBE2I promoter methylation and protein levels were performed using the UALCAN database. The underlying mechanisms of UBE2I involvement in pan-DSTs were visualized using interaction networks. The diagnostic value of UBE2I in pan-DSTs was identified using the Oncomine database. Results. UBE2I was differentially and highly expressed in cholangiocarcinoma (CHOL), pancreatic adenocarcinoma (PAAD), colon adenocarcinoma (COAD), rectal adenocarcinoma (READ), liver hepatocellular carcinoma (LIHC), and stomach adenocarcinoma (STAD). According to survival analysis, upregulated UBE2I was associated with adverse overall and disease-free survival in PAAD and favorable overall survival in READ. UBE2I expression was partially linked to the purity of immune infiltration in COAD, LIHC, PAAD, READ, and STAD, as indicated by the immune infiltration analysis. Promoter methylation analysis showed differential and high methylation of UBE2I in PAAD as well as stratified analysis by gender, nodal metastasis, and race. Protein expression analysis in colon cancer revealed that UBE2I had differential and high expression in tumors as well as stratified analysis by gender, tumor histology, race, and tumor stage. Mechanism explorations demonstrated that in COAD and PAAD, UBE2I was involved in spliceosomal snRNP complex, Notch signaling pathway, etc. Diagnostic analysis indicated that UBE2I had consistent diagnostic value for COAD and PAAD. Conclusions. Upregulated UBE2I may be a diagnostic and surveillance predictive signature for PAAD and COAD. The potential significance of immune infiltrates and promoter methylation in PAAD and COAD needs further exploration.
APA, Harvard, Vancouver, ISO, and other styles
36

Fang, Xiaona, Shan Liu, Liuxian Ban, Jiao Huang, Jie Luo, Lanqi Gong, Baifeng Zhang, et al. "Abstract 2441: Identification and characterization of the role of KRT17 in the stemness regulation of hepatocellular carcinoma." Cancer Research 83, no. 7_Supplement (April 4, 2023): 2441. http://dx.doi.org/10.1158/1538-7445.am2023-2441.

Full text
Abstract:
Abstract Hepatocellular carcinoma (HCC) ranks as the fifth most common cancer and the second leading cause of cancer-related mortality worldwide. The overall prognosis of HCC patients is very poor because it is susceptible to recurrence and metastasis. According to the cancer stem cell (CSC) model, there is a small subset of cancer cells within the tumor bulk that is responsible for tumor relapse and metastasis. CSCs represent cells that can self-renew, initiate tumors, and keep resistant to anti-cancer treatment. However, the fate and signatures of CSCs in HCC remain elusive. Our previous in vitro hepatocyte differentiation model successfully mimicked the liver developmental process from the stage of embryonic stem cells, endoderm, liver progenitor cells, and premature hepatocytes, together with one pair of nontumor and tumor tissue from an HCC patient. We found that the expression of keratin 17 (KRT17) reached its peak at the liver progenitor stage and was down-regulated along with liver development, which shared the same expression pattern with keratin 19 (CK19), a well-known HCC stemness-associated gene. Single-cell RNA sequencing in 9 HCC clinical samples from the GEO dataset revealed that KRT17 was expressed in less than 1% of tumor cells. Moreover, KRT17 shared similar expression patterns among hepatocytes, non-stem like HCC and stem like HCC subpopulations with a panel of hepatic stem cell markers and showed reverse trends with a list of hepatic markers. Clinically, KRT17 was highly expressed in cancer tissues compared to normal counterparts and positively correlated with poor overall survival. Functional assays indicated that KRT17 could considerably facilitate the self-renewal, growth, and metastasis properties of HCC both in vitro and in vivo. Also, KRT17 endowed HCC cells with enhanced resistance to sorafenib treatment. Moreover, the expressions of the CSC marker (CD133) and oncofetal drivers (NANOG and KLF4) were significantly upregulated upon KRT17 overexpression. Bioinformatic analysis using gene expression profiles of patients from the TCGA database revealed that development- and differentiation-related biological processes were dramatically enriched in KRT17-high patients. The underlying molecular mechanism of how KRT17 augments HCC stemness still needs further study. Overall, our study uncovered a new connection between KRT17 and HCC stemness and progression. KRT17 might be a novel marker of CSC and a prognostic biomarker for the treatment of HCC patients. Citation Format: Xiaona Fang, Shan Liu, Liuxian Ban, Jiao Huang, Jie Luo, Lanqi Gong, Baifeng Zhang, Beilei Liu, Jinlin Huang, Yuma Yang, Ching Ngar Wong, Qian Yan, Xin-Yuan Guan. Identification and characterization of the role of KRT17 in the stemness regulation of hepatocellular carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2441.
APA, Harvard, Vancouver, ISO, and other styles
37

Sakamoto, Yuzuru, Sachiyo Yoshio, Akihisa Nagatsu, Yoh Asahi, Shingo Shimada, Tatsuya Orimo, Hirofumi Kamachi, Toshiya Kamiyama, Tatsuya Kanto, and Akinobu Taketomi. "Increased frequency of PD-1+CD57+Siglec-7- dysfunctional NK cells in patients with nonalcoholic fatty liver disease." Journal of Clinical Oncology 38, no. 4_suppl (February 1, 2020): 589. http://dx.doi.org/10.1200/jco.2020.38.4_suppl.589.

Full text
Abstract:
589 Background: The proportion of non-alcoholic fatty liver disease (NAFLD) has been increasing as a cause of hepatocellular carcinoma (HCC) worldwide. Natural killer (NK) cells are involved in the first line of immune defense against cancer. NK cell function is regulated by activating and inhibitory NK cell receptors. However, the role of NK cells in the pathogenesis of NAFLD and NAFLD-HCC is still largely unknown. In this study, we aimed to clarify the phenotypic and functional features of NK cells in NAFLD and NAFLD-HCC patients. Methods: We performed mass cytometry (CyTOF) and flow cytometry analysis of NK cells in 33 NAFLD patients (22 chronic hepatitis (CH), 8 liver cirrhosis (LC), 3 with HCC (HCC)) and 9 healthy donors (HDs). We compared surface markers on NK cells in cancerous and non-cancerous intrahepatic lymphocytes (IHLs). We also measured NK cell function in the presence of IL-12 and IL-18. Results: The frequency of NK cells was lower in NAFLD patients compared to HDs. PD-1, CD57, ILT2 were highly expressed, and Siglec-7, NKp30, NKp46 were downregulated on NK cells from NAFLD patients, compared with those of HDs. In NAFLD patients, Siglec-7 levels on NK cells were negatively correlated with PD-1 and CD57, and positively correlated with NKp30 and NKp46. The other inhibitory markers (NKG2A, KIR3DL1 and KIRDL2/L3), activating markers (CD69 and NKG2D) and checkpoint markers (Tim-3 and TIGIT) were comparable between NAFLD patients and HDs. PD-1 and CD57 expression levels on NK cells were also significantly upregulated in NAFLD-HCC patients than those in HDs. CD57 was rarely expressed on NK cells in non-cancerous IHLs, on the other hand, highly expressed in cancerous IHLs. The IFNγ production and CD107a expression on NK cells were also decreased in NAFLD patients. PD-1+CD57+Siglec-7− NK cells were observed in NAFLD patients, rarely in HDs. PD-1+CD57+Siglec-7− NK cells were functionally impaired compared to other NK subsets. Conclusions: In patients with NAFLD, NK cells are reduced and functionally impaired, the reason of which may be the increased proportion of dysfunctional PD-1+CD57+Siglec-7− NK subset, and dysfunctional NK cells might be related to impairment of surveillance for HCC.
APA, Harvard, Vancouver, ISO, and other styles
38

Beaumont, Jamie, Eric Aboagye, Beata Wojciak-Stothard, Gilberto Serrano-De-Almeida, Robert Glen, and Rohini Sharma. "Abstract 5398: Apelinergic signalling in hepatocellular carcinoma (HCC): A new therapeutic treatment option." Cancer Research 82, no. 12_Supplement (June 15, 2022): 5398. http://dx.doi.org/10.1158/1538-7445.am2022-5398.

Full text
Abstract:
Abstract HCC is the third most frequent cancer-related death worldwide and incidence continues to rise. Most patients presenting with advanced stage disease where their treatment goal is palliation, emphasizing the requirement for new treatment options. HCC is characterized by arterialization of its blood supply deemed pathognomonic for its development. We investigated the neoangiogenic peptide apelin as a driver of carcinogenesis in HCC followed by the efficacy of novel apelin antagonists as a therapeutic strategy in-vitro. Firstly, meta-analysis conducted through the TCGA database showed that apelin is significantly upregulated in liver cancer compared to healthy tissue and this correlated with a worse mean overall survival. Additionally, enzyme-linked immunosorbent assay (ELISA) of patient serum samples corroborated that apelin concentration significantly increases from healthy liver to cirrhosis and then further through tumorigenesis. In-vitro studies across multiple hepatocyte cell lines in media conditioned with elevated serum apelin levels were then performed to investigate the effects of increased apelin. All cell-lines demonstrated an increase in migration and proliferative properties with naturally higher basal apelin expressing cell lines showing the most significant changes. Using knockdown models, we elucidated hypoxia a well-known driver of angiogenesis, up-regulates apelin expression through a HIF-1α dependent mechanism. Furthermore, increased apelin expression resulted in upregulation of its G-protein coupled receptor, APJ which initiates its downstream signaling pathways, together referred to as apelinergic signaling, demonstrating an autocrine loop. Co-culture of hepatocytes with hepatic stellate cells (HSCs), activated in cirrhosis, further elucidated a paracrine signaling loop whereby secreted chemokines from HSCs, which including apelin, enhanced hepatocyte proliferation and migration. Assessing apelin as a therapeutic target, several novel antagonists were designed in-silico and investigated across multiple hepatocyte cell lines. MM315 showed the most promise with significant cytostatic properties at nanomolar levels coupled with a high binding affinity to APJ at picomolar levels demonstrated through blocking studies. As HCC is a highly vascularized cancer, we explored the likely effects of these antagonists on endothelial cells, cultured in the presence of IC50 concentrations of each antagonist. MM315 treatment resulted in a significant reduction of abnormal blood vessel formation. Overall, our results suggest inhibiting apelinergic signaling is a promising therapeutic treatment for HCC, with MM315 being a lead compound. In-vivo work which will be presented. Citation Format: Jamie Beaumont, Eric Aboagye, Beata Wojciak-Stothard, Gilberto Serrano-De-Almeida, Robert Glen, Rohini Sharma. Apelinergic signalling in hepatocellular carcinoma (HCC): A new therapeutic treatment option [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5398.
APA, Harvard, Vancouver, ISO, and other styles
39

Liu, Jun, Zheng Chen, and Wenli Li. "Machine Learning for Building Immune Genetic Model in Hepatocellular Carcinoma Patients." Journal of Oncology 2021 (March 17, 2021): 1–15. http://dx.doi.org/10.1155/2021/6676537.

Full text
Abstract:
Background. Hepatocellular carcinoma (HCC) is the leading liver cancer with special immune microenvironment, which played vital roles in tumor relapse and poor drug responses. In this study, we aimed to explore the prognostic immune signatures in HCC and tried to construct an immune-risk model for patient evaluation. Methods. RNA sequencing profiles of HCC patients were collected from the cancer genome Atlas (TCGA), international cancer genome consortium (ICGC), and gene expression omnibus (GEO) databases (GSE14520). Differentially expressed immune genes, derived from ImmPort database and MSigDB signaling pathway lists, between tumor and normal tissues were analyzed with Limma package in R environment. Univariate Cox regression was performed to find survival-related immune genes in TCGA dataset, and in further random forest algorithm analysis, significantly changed immune genes were used to generate a multivariate Cox model to calculate the corresponding immune-risk score. The model was examined in the other two datasets with recipient operation curve (ROC) and survival analysis. Risk effects of immune-risk score and clinical characteristics of patients were individually evaluated, and significant factors were then used to generate a nomogram. Results. There were 52 downregulated and 259 upregulated immune genes between tumor and relatively normal tissues, and the final immune-risk model (based on SPP1, BRD8, NDRG1, KITLG, HSPA4, TRAF3, ITGAV and MAP4K2) can better differentiate patients into high and low immune-risk subpopulations, in which high score patients showed worse outcomes after resection ( p < 0.05 ). The differentially enriched pathways between the two groups were mainly about cell proliferation and cytokine production, and calculated immune-risk score was also highly correlated with immune infiltration levels. The nomogram, constructed with immune-risk score and tumor stages, showed high accuracy and clinical benefits in prediction of 1-, 3- and 5-year overall survival, which is useful in clinical practice. Conclusion. The immune-risk model, based on expression of SPP1, BRD8, NDRG1, KITLG, HSPA4, TRAF3, ITGAV, and MAP4K2, can better differentiate patients into high and low immune-risk groups. Combined nomogram, using immune-risk score and tumor stages, could make accurate prediction of 1-, 3- and 5-year survival in HCC patients.
APA, Harvard, Vancouver, ISO, and other styles
40

Jung, Ji Hoon, Hyo-Jung Lee, Ju-Ha Kim, Deok Yong Sim, Eunji Im, Sinae Kim, Suhwan Chang, and Sung-Hoon Kim. "Colocalization of MID1IP1 and c-Myc is Critically Involved in Liver Cancer Growth via Regulation of Ribosomal Protein L5 and L11 and CNOT2." Cells 9, no. 4 (April 16, 2020): 985. http://dx.doi.org/10.3390/cells9040985.

Full text
Abstract:
Though midline1 interacting protein 1 (MID1IP1) was known as one of the glucose-responsive genes regulated by carbohydrate response element binding protein (ChREBP), the underlying mechanisms for its oncogenic role were never explored. Thus, in the present study, the underlying molecular mechanism of MID1P1 was elucidated mainly in HepG2 and Huh7 hepatocellular carcinoma cells (HCCs). MID1IP1 was highly expressed in HepG2, Huh7, SK-Hep1, PLC/PRF5, and immortalized hepatocyte LX-2 cells more than in normal hepatocyte AML-12 cells. MID1IP1 depletion reduced the viability and the number of colonies and also increased sub G1 population and the number of TUNEL-positive cells in HepG2 and Huh7 cells. Consistently, MID1IP1 depletion attenuated pro-poly (ADP-ribose) polymerase (pro-PARP), c-Myc and activated p21, while MID1IP1 overexpression activated c-Myc and reduced p21. Furthermore, MID1IP1 depletion synergistically attenuated c-Myc stability in HepG2 and Huh7 cells. Of note, MID1IP1 depletion upregulated the expression of ribosomal protein L5 or L11, while loss of L5 or L11 rescued c-Myc in MID1IP1 depleted HepG2 and Huh7 cells. Interestingly, tissue array showed that the overexpression of MID1IP1 was colocalized with c-Myc in human HCC tissues, which was verified in HepG2 and Huh7 cells by Immunofluorescence. Notably, depletion of CCR4-NOT2 (CNOT2) with adipogenic activity enhanced the antitumor effect of MID1IP1 depletion to reduce c-Myc, procaspase 3 and pro-PARP in HepG2, Huh7 and HCT116 cells. Overall, these findings provide novel insight that MID1IP1 promotes the growth of liver cancer via colocalization with c-Myc mediated by ribosomal proteins L5 and L11 and CNOT2 as a potent oncogenic molecule.
APA, Harvard, Vancouver, ISO, and other styles
41

Zhang, Hailong. "Abstract B031: PRC2 inhibitor suppresses angiogenesis in Hepatocellular carcinoma (HCC) by up-regulation of anti-angiogenic factors vasohibin1 (VASH1) and tissue inhibitor of metalloproteinases 3 (TIMP-3)." Cancer Research 82, no. 23_Supplement_2 (December 1, 2022): B031. http://dx.doi.org/10.1158/1538-7445.cancepi22-b031.

Full text
Abstract:
Abstract Tumor-associated angiogenesis plays a crucial role in the progression of tumor, especially in highly vascularized cancer, such as Liver hepatocellular carcinoma (LIHC), ovarian cancer, lung cancer, human head and neck cancers. Clinical research has shown that LIHC cells upregulate expression of vascular endothelial growth factor A (VEGFA), and induce proliferation and migration of epithelial cells under hypoxia condition, however, the mechanism of angiogenesis has not been completely elucidated in LIHC, especially in epigenetic regulation level. Polycomb repressive complex 2 (PRC2) is a multi-subunit protein complex that catalyzes the methylation of histone H3 at lysine 27, Enhancer of Zeste homolog 2 (EZH2) is the catalytic subunit, while embryonic ectoderm development (EED) recognizes H3K27me3 and enhance the catalytic activity of EZH2. A recent study suggests PRC2 promote angiogenesis in human ovarian carcinoma, while the detailed mechanism this process has not been illustrated. In this paper, we show BR1763, a potent, selective and orally bioavailable small-molecule inhibitor of EED, inhibits PRC2 activity, promote expression of vasohibin1 (VASH1) and tissue inhibitor of metalloproteinases 3 (TIMP3) in certain Hepatocellular Carcinoma (HCC) cells, and induces tumor growth inhibition by suppression of tumor-associated angiogenesis in vivo. Citation Format: Hailong Zhang. PRC2 inhibitor suppresses angiogenesis in Hepatocellular carcinoma (HCC) by up-regulation of anti-angiogenic factors vasohibin1 (VASH1) and tissue inhibitor of metalloproteinases 3 (TIMP-3). [abstract]. In: Proceedings of the AACR Special Conference: Cancer Epigenomics; 2022 Oct 6-8; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2022;82(23 Suppl_2):Abstract nr B031.
APA, Harvard, Vancouver, ISO, and other styles
42

Vijay-Kumar, Matam, Beng San Yeoh, Vishal Singh, Rachel M. Golonka, and Piu Saha. "Dietary Soluble Fiber Induces HCC in Dysbiotic Mice Through a Spectrum of Immunosuppressive Mediators." Journal of Immunology 202, no. 1_Supplement (May 1, 2019): 59.3. http://dx.doi.org/10.4049/jimmunol.202.supp.59.3.

Full text
Abstract:
Abstract Dietary fermentable, soluble fibers (e.g. inulin) are known for promoting health; yet, ~40% of Toll-like receptor 5 deficient (T5KO) mice fed inulin-containing diet (ICD) develop icteric hepatocellular carcinoma (HCC) (Cell, 2018). Specifically, the disease progresses starting with hepatic cholestasis and neutrophilic inflammation at one month, followed by HCC at 6 months of ICD feeding. This diet-driven HCC was dependent on the gut microbiota and was mediated, largely in part, via surfeit production of immunosuppressive short-chain fatty acids (SCFA) and secondary bile acids, presumably as compensatory mechanisms against inflammation in the 40% pre-HCC T5KO mice. What was most striking was the upregulation of systemic bilirubin (a host immunosuppressive metabolite) within one week of ICD feeding. The hyperbilirubinemia was accompanied with an increase in IgA by ~50 and ~10-fold in the serum and liver, respectively, compared to the 60% non-HCC T5KO mice. Markers for anti-tumor T cell exhaustion, i.e. hepatic PD-L1 and layilin, were also upregulated, where the former was elevated within a week of feeding and persisted, while the latter was observed at 6 months. Hepatic immune cell phenotyping after the onset of HCC revealed substantial increase in CD4+FOXP3+PD-L1+T cells and IgA+IL10+PD-L1+Bcells, with the latter denoting a recently identified subset of highly immunosuppressive B cells. Our unprecedented findings collectively demonstrate that dietary fibers and SCFA —which are widely-held to be anti-inflammatory— could potentiate immunosuppression and impede anti-tumor immune surveillance in the tumor microenvironment in mice with gut microbiota dysbiosis.
APA, Harvard, Vancouver, ISO, and other styles
43

Duffy, Austin G., Osama E. Rahma, Susanna Varkey Ulahannan, Suzanne Fioravanti, Aradhana Venkatesan, Ismail B. Turkbey, Peter L. Choyke, Jane B. Trepel, William Douglas Figg, and Tim F. Greten. "A phase II study of TR105 in patients with hepatocellular carcinoma (HCC) who have progressed on sorafenib." Journal of Clinical Oncology 31, no. 15_suppl (May 20, 2013): e15177-e15177. http://dx.doi.org/10.1200/jco.2013.31.15_suppl.e15177.

Full text
Abstract:
e15177 Background: CD105, also known as endoglin, is an endothelial cell membrane receptor that is highly expressed on tumor vasculature, including HCC. CD105 is essential for angiogenesis and its expression is upregulated by hypoxia and VEGF inhibition. TRC105 is a chimeric IgG1 anti-CD105 monoclonal antibody that inhibits angiogenesis and tumor growth by endothelial cell growth inhibition, ADCC and apoptosis. Methods: Patients with HCC and compensated liver function (Childs Pugh A/B7), ECOG 0/1, were enrolled to a single arm phase II study of TRC105 15mg/kg IV every 2 weeks. Patients must have progressed on or been intolerant of prior sorafenib. A Simon optimal 2-stage design was employed with a 50% 4-month PFS target for progression to the second stage. Correlative biomarkers evaluated included: DCE-MRI; FDG-PET; color Doppler ultrasonography; circulating endothelial progenitor cells, plasma levels of angiogenic factors; soluble CD105 and tumor IHC for CD105. Immunogenicity studies were also performed. Results: 8 pts have been treated so far; M:F 6:2; Median age = 58 (range 24-67); 7 pts had progressed following sorafenib (N=1 intolerant). N=1 pt had fibrolammellar HCC. There were no grade 3/4 treatment-related toxicities. Most frequent toxicities were headache (G2; N=3) and epistaxis (G1; N=4). One patient with a history of ischemic heart disease developed acute cardiac syndrome during the first infusion and was replaced. No patient was progression free at 4 months by RECIST criteria. Median time on study was 8 weeks (range 4-16 weeks). Median no. of doses administered was 4 (range 2-7). Preliminary evidence of biologic response was seen on DCE-MRI in 1pt (of 3 evaluable) with a reduction in kTrans and kep and 3pts (of 3 evaluable) who demonstrated reduction in intra-tumoral color flow on color Doppler. Conclusions: TRC105 is well tolerated in this HCC population post-sorafenib. Although there was biological evidence of antiangiogenic activity it is unlikely that the study will proceed to the second stage. Full clinical and correlative data for the first stage of the study will be presented. Future role of TRC105 in HCC will most likely be as combination therapy with sorafenib or other anti-VEGF therapy. Clinical trial information: NCT01375569.
APA, Harvard, Vancouver, ISO, and other styles
44

Kwon, Young-Chan, Sandip K. Bose, Robert Steele, Keith Meyer, Adrian M. Di Bisceglie, Ratna B. Ray, and Ranjit Ray. "Promotion of Cancer Stem-Like Cell Properties in Hepatitis C Virus-Infected Hepatocytes." Journal of Virology 89, no. 22 (September 9, 2015): 11549–56. http://dx.doi.org/10.1128/jvi.01946-15.

Full text
Abstract:
ABSTRACTWe have previously reported that hepatitis C virus (HCV) infection of primary human hepatocytes (PHH) induces the epithelial mesenchymal transition (EMT) state and extends hepatocyte life span (S. K. Bose, K. Meyer, A. M. Di Bisceglie, R. B. Ray, and R. Ray, J Virol 86:13621–13628, 2012,http://dx.doi.org/10.1128/JVI.02016-12). These hepatocytes displayed sphere formation on ultralow binding plates and survived for more than 12 weeks. The sphere-forming hepatocytes expressed a number of cancer stem-like cell (CSC) markers, including high levels of the stem cell factor receptor c-Kit. The c-Kit receptor is regarded as one of the CSC markers in hepatocellular carcinoma (HCC). Analysis of c-Kit mRNA displayed a significant increase in the liver biopsy specimens of chronically HCV-infected patients. We also found c-Kit is highly expressed in transformed human hepatocytes (THH) infectedin vitrowith cell culture-grown HCV genotype 2a. Further studies suggested that HCV core protein significantly upregulates c-Kit expression at the transcriptional level. HCV infection of THH led to a significant increase in the number of spheres displayed on ultralow binding plates and in enhanced EMT and CSC markers and tumor growth in immunodeficient mice. The use of imatinib or dasatinib as a c-Kit inhibitor reduced the level of sphere-forming cells in culture. The sphere-forming cells were sensitive to treatment with sorafenib, a multikinase inhibitor, that is used for HCC treatment. Further, stattic, an inhibitor of the Stat3 molecule, induced sphere-forming cell death. A combination of sorafenib and stattic had a significantly stronger effect, leading to cell death. These results suggested that HCV infection potentiates CSC generation, and selected drugs can be targeted to efficiently inhibit cell growth.IMPORTANCEHCV infection may develop into HCC as an end-stage liver disease. We focused on understanding the mechanism for the risk of HCC from chronic HCV infection and identified targets for treatment. HCV-infected primary and transformed human hepatocytes (PHH or THH) generated CSC. HCV-induced spheres were highly sensitive to cell death from sorafenib and stattic treatment. Thus, our study is highly significant for HCV-associated HCC, with the potential for developing a target-specific strategy for improved therapies.
APA, Harvard, Vancouver, ISO, and other styles
45

Verslype, Chris, Hannah van Malenstein, Jeroen Dekervel, Petra Windmolders, Louis Libbrecht, Rudy van Eijsden, Frederik Nevens, and Jos van Pelt. "Resistance development after long-term sorafenib exposure in hepatocellular cancer cell lines and risk of rebound growth and epithelial to mesenchymal transition." Journal of Clinical Oncology 30, no. 4_suppl (February 1, 2012): 216. http://dx.doi.org/10.1200/jco.2012.30.4_suppl.216.

Full text
Abstract:
216 Background: Sorafenib, a multi tyrosine kinase inhibitor, is the first line treatment in patients with advanced hepatocellular carcinoma (HCC). It leads to a survival benefit but treatment with sorafenib is hampered by two phenomena: patients can develop important side-effects and eventually all patients show progression. We aimed to determine the effects of long-term exposure to sorafenib and its withdrawal in vitro. Methods: We developed sorafenib resistant liver cancer cell lines (HepG2, WRL-68 and Huh-7) by slowly increasing sorafenib concentrations. XTT- and BrdU-assay were used to study the effect of sorafenib withdrawal on proliferation and metabolism. Morphological changes were examined with immuocytochemistry, gene expression changes with RT-PCR and the invasive potential with matrigel invasion chambers. Microarray was performed on resistant HepG2 cells. Results: HepG2 cells (6µM), WRL-68 cells (6µM) and Huh-7 cells (5µM) became resistant to sorafenib. Resistance was confirmed with a shift of the IC50 to ±16µM and ongoing phosphorylation of ERK during sorafenib exposure. All three resistant cell types showed significant increased proliferation and metabolic activity after withdrawal of sorafenib. The HepG2 resistant cells have undergone an epithelial to mesenchymal transition (EMT) with loss of E-cadherin and high expression of vimentin. The cells that displayed EMT became spindle shaped and were highly invasive. Furthermore, gene expression profiling confirmed EMT changes in a large set of EMT-related genes. Considering drug metabolism, the HepG2 sorafenib resistant cells showed a downregulation of UDP glucuronosyltransferases (UGT) and cytochromes P450, such as CYP3A4. There was a strong downregulation of different ABC-transporters, although breast cancer resistance protein (ABCG2) was upregulated. Conclusions: Long-term treatment with sorafenib can lead to the development of resistant cells, even with an aggressive phenotype because the cells can undergo EMT. Furthermore we demonstrated that abrogation of treatment leads to rebound growth, suggesting the importance of aggressive management of side-effects.
APA, Harvard, Vancouver, ISO, and other styles
46

Weinberger, Paul, Sithara Raju Ponny, Hongyan Xu, Shan Bai, Robert Smallridge, John Copland, and Ashok Sharma. "Cell Cycle M-Phase Genes Are Highly Upregulated in Anaplastic Thyroid Carcinoma." Thyroid 27, no. 2 (February 2017): 236–52. http://dx.doi.org/10.1089/thy.2016.0285.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Paganoni, Rossana, André Lechel, and Maja Vujic Spasic. "Iron at the Interface of Hepatocellular Carcinoma." International Journal of Molecular Sciences 22, no. 8 (April 15, 2021): 4097. http://dx.doi.org/10.3390/ijms22084097.

Full text
Abstract:
Cancer incidence and mortality are rapidly growing, with liver cancer being the sixth most diagnosed cancer worldwide and the third leading cause of cancer death in 2020. A number of risk factors have been identified that trigger the progression to hepatocellular carcinoma. In this review, we focus on iron as a potential risk factor for liver carcinogenesis. Molecules involved in the regulation of iron metabolism are often upregulated in cancer cells, in order to provide a supply of this essential trace element for all stages of tumor development, survival, proliferation, and metastasis. Thus, cellular and systemic iron levels must be tightly regulated to prevent or delay liver cancer progression. Disorders associated with dysregulated iron metabolism are characterized with increased susceptibility to hepatocellular carcinoma. This review discusses the association of iron with metabolic disorders such as hereditary hemochromatosis, non-alcoholic fatty liver disease, obesity, and type 2 diabetes, in the background of hepatocellular carcinoma.
APA, Harvard, Vancouver, ISO, and other styles
48

Liang, Zenghui, Yingtang Gao, Wenxia Shi, Daokuan Zhai, Shilei Li, Li Jing, Hua Guo, Tong Liu, Yajie Wang, and Zhi Du. "Expression and Significance of MicroRNA-183 in Hepatocellular Carcinoma." Scientific World Journal 2013 (2013): 1–6. http://dx.doi.org/10.1155/2013/381874.

Full text
Abstract:
Objective. In our previous study, we found that some miRNAs were deregulated in hepatocellular carcinoma (HCC), including miR-183. However, the expression of miR-183 in the progression of benign liver diseases to HCC and its correlation with clinicopathologic factors remain undefined.Methods. MiR-183 expression was measured in normal controls (NC) (n=21), chronic viral hepatitis B or C (CH) tissues (n=10), liver cirrhosis (LC) tissues (n=18), HCC tissues (n=92), and adjacent nontumor tissues (NT) (n=92) by quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR).Results. The expression levels of miR-183 were significantly higher in HCC than in NT, LC, CH, and NL (P=0.001,P<0.001,P=0.011,P<0.001, resp.). The upregulated miR-183 in HCC was correlated with TNM stage (P=0.042) and cirrhosis (P=0.025). The Kaplan-Meier survival analysis showed that miR-183 expression was not associated with the survival of HCC patients. However, miR-183 yielded an area under the curve (AUC) of 0.808 with 59.8% sensitivity and 91.8% specificity in discriminating HCC from benign liver diseases (CH and LC) or NC.Conclusions. The upregulated miR-183 may associate with onset and progression of HCC, but not with the patient survival. A further research is needed to determine the potential of miR-183 as biomarker for HCC.
APA, Harvard, Vancouver, ISO, and other styles
49

Matouk, Imad J., Ibrahim Abbasi, Abraham Hochberg, Eithan Galun, Hassan Dweik, and Mutaz Akkawi. "Highly upregulated in liver cancer noncoding RNA is overexpressed in hepatic colorectal metastasis." European Journal of Gastroenterology & Hepatology 21, no. 6 (June 2009): 688–92. http://dx.doi.org/10.1097/meg.0b013e328306a3a2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
50

Ghafouri‐Fard, Soudeh, Mohammadhosein Esmaeili, Mohammad Taheri, and Majid Samsami. "Highly upregulated in liver cancer (HULC): An update on its role in carcinogenesis." Journal of Cellular Physiology 235, no. 12 (May 5, 2020): 9071–79. http://dx.doi.org/10.1002/jcp.29765.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography