Catenibacteriummitsuokai promotes hepatocellular carcinogenesis by binding to hepatocytes and generating quinolinic acid.
Catenibacteriummitsuokai promotes hepatocellular carcinogenesis by binding to hepatocytes and generating quinolinic acid.
51
- 10.1038/s43018-021-00251-3
- Sep 27, 2021
- Nature Cancer
23
- 10.1016/j.ymben.2021.06.007
- Jun 27, 2021
- Metabolic Engineering
26
- 10.1007/978-3-319-09614-8_10
- Nov 10, 2014
- Advances in experimental medicine and biology
787
- 10.1016/j.tibs.2009.10.002
- Oct 31, 2009
- Trends in Biochemical Sciences
87
- 10.1136/gutjnl-2021-325189
- Aug 3, 2021
- Gut
83
- 10.1038/mi.2015.27
- Apr 29, 2015
- Mucosal Immunology
1680
- 10.1126/science.aay9189
- May 28, 2020
- Science
62
- 10.1038/s41598-020-78141-4
- Dec 1, 2020
- Scientific Reports
2
- 10.1186/s12885-024-13161-1
- Nov 29, 2024
- BMC Cancer
2
- 10.1186/s13099-024-00650-8
- Oct 5, 2024
- Gut Pathogens
- Research Article
- 10.1158/1538-7445.transcontrol16-a18
- Mar 14, 2017
- Cancer Research
Background and Significance: Non-alcoholic fatty liver disease (NAFLD) is a major risk factor for the development of hepatocellular carcinoma (HCC). Even with successful surgical removal, the presence of NAFLD is associated with an increased recurrence of HCC. Despite the apparent association between NAFLD and HCC, the mechanistic pathways that link both disorders are essentially unknown and therapeutic agents for both disorders are lacking. MicroRNAs (miRNAs) play critical roles in both cancer and metabolic disorders, and present tremendous therapeutic potential. Methods: We used four mouse models including ATK/Ras HCC mice, cMyc HCC mice, xenograft HCC mice and dietary obese mice to evaluate the therapeutic effects of miR-206 on the pathogenesis of HCC and NAFLD. Mini-circle and Sleeping Beauty system were used to deliver miR-206 into livers of mice. Results: miRNA profiling revealed that hepatic fat accumulation impaired biogenesis of miR-206 that is also robustly under-expressed in livers of HCC mice and HCC patients. System administration of miR-206 completely prevented HCC development in both cMyc and AKT/Ras HCC mice, while 100% control mice died from lethal tumor burdens. Upon dissection, no tumor nodules were observed in livers of both AKT/Ras and cMyc mice treated with miR-206. Notably, liver-specific expression of miR-206 displayed the robust therapeutic effects on obesity and NAFLD in dietary obese mice and delivery of miR-206 into human hepatocytes significantly reduced intracellular lipid content. Combining bioinformatic prediction and molecular and cellular approaches, we identified cMet (Met proto-oncogene), CCND1, CDK4/6, EGFR, and PTPN1 (protein tyrosine phosphatase 1B) as essential direct targets of miR-206. It is known that PTPN1 can dephosphorylate PP2A (phosphatase 2A). Dephosphorylated PP2A (active form) can activate transcription of Srebp1c by dephosphorylating Sp1, a transcription activator of Srebp1c. As expected, through modulating PTPN1-PP2A-SP1-Srebp1c axis, miR-206 impaired Srebp1c-mediated lipogenesis in livers of dietary obese mice and in human hepatocytes treated with a fatty acid, while the prevention of the interaction between miR-206 and PTPN1 offset the inhibitory effects of miR-206 on lipogenesis and hepatic lipid accumulation in vivo and in vitro, suggesting that PTPN1 mediates the therapeutic effects of miR-206 on NAFLD. Through inhibiting expression of cMet, CCND1, CDK4/6, and EGFR, miR-206 completely prevented colony formation of HCC cell lines with divergent backgrounds and growth of xenograft HCC tumor from different HCC cells by impairing cMet signaling, EGFR signaling and cell cycle progression. Conclusion: This defines a critical role of miR-206 in preventing the pathogenesis of highly-associated NAFLD and HCC, and suggests its strong potential as a therapeutic strategy for both disorders. Citation Format: Guisheng Song, Heng Wu, Junyan Tao, Xin Chen. microRNA-206 as a potential therapeutic approach for highly-associated NAFLD and HCC. [abstract]. In: Proceedings of the AACR Special Conference on Translational Control of Cancer: A New Frontier in Cancer Biology and Therapy; 2016 Oct 27-30; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2017;77(6 Suppl):Abstract nr A18.
- Research Article
45
- 10.1016/j.omtn.2019.08.020
- Aug 28, 2019
- Molecular Therapy Nucleic Acids
lncRNA MAGI2-AS3 Prevents the Development of HCC via Recruiting KDM1A and Promoting H3K4me2 Demethylation of the RACGAP1 Promoter
- Research Article
113
- 10.1186/s13046-019-1188-x
- May 14, 2019
- Journal of Experimental & Clinical Cancer Research
BackgroundAccumulating evidence has highlighted the potential role of long non-coding RNAs (lncRNAs) in the biological behaviors of hepatocellular carcinoma (HCC). Here, we elucidated the function and possible molecular mechanisms of the effect of lncRNA-AGAP2-AS1 on the biological behaviors of HCC.MethodsEdU, Transwell and flow cytometry were used to determine proliferation, migration, invasion and apoptosis of HCC cells in vitro. The subcutaneous tumor model and lung metastasis mouse model in nude mice was established to detect tumor growth and metastasis of HCC in vivo. The direct binding of miR-16-5p to 3’UTR of ANXA11 was confirmed by luciferase reporter assay. The expression of AGAP2-AS1 and miR-16-5p in HCC specimens and cell lines were detected by real-time PCR. The correlation among AGAP2-AS1 and miR-16-5p were disclosed by a dual-luciferase reporter assay, RIP assay and biotin pull-down assay.ResultsHere, we demonstrated that AGAP2-AS1 expression was up-regulated in HCC tissues and cell lines, especially in metastatic and recurrent cases. Gain- and loss-of-function experiments indicated that AGAP2-AS1 promoted cell proliferation, migration, invasion, EMT progression and inhibited apoptosis of HCC cells in vitro and in vivo. Further studies demonstrated that AGAP2-AS1 could function as a competing endogenous RNA (ceRNA) by sponging miR-16-5p in HCC cells. Functionally, gain- and loss-of-function studies showed that miR-16-5p promoted HCC progression and alteration of miR-16-5p abolished the promotive effects of AGAP2-AS1 on HCC cells. Moreover, ANXA11 was identified as direct downstream targets of miR-16-5p in HCC cells, and mediated the functional effects of miR-16-5p and AGAP2-AS1 in HCC, resulting in AKT signaling activation. Clinically, AGAP2-AS1 and miR-16-5p expression were markedly correlated with adverse clinical features and poor prognosis of HCC patients. We showed that hypoxia was responsible for the overexpression of AGAP2-AS1 in HCC. And the promoting effects of hypoxia on metastasis and EMT of HCC cells were reversed by AGAP2-AS1 knockdown.ConclusionsTaken together, this research supports the first evidence that AGAP2-AS1 plays an oncogenic role in HCC via AGAP2-AS1/miR-16-5p/ANXA11/AKT axis pathway and represents a promising therapeutic strategy for HCC patients.
- Research Article
54
- 10.2353/ajpath.2010.090356
- Mar 1, 2010
- The American Journal of Pathology
Reduced Expression of Fibroblast Growth Factor Receptor 2IIIb in Hepatocellular Carcinoma Induces a More Aggressive Growth
- Research Article
12
- 10.18632/aging.103552
- Jul 20, 2020
- Aging
Objective: An increasing number of studies have indicated that long noncoding RNAs (lncRNAs) play an important role in the pathogenesis of hepatocellular carcinoma (HCC). In this study, we aimed to clarify the roles of RP11-295G20.2 in HCC progression and the underlying molecular mechanisms.Results: Bioinformatics analyses based TCGA data suggested that RP11-295G20.2 was significantly upregulated in HCC tissues and increased RP11-295G20.2 expression level correlated with poor overall survival of patients with HCC. The results of RT-PCR further showed that RP11-295G20.2 was upregulated in HCC tissues and cell lines. Functionally, RP11-295G20.2 knockdown significantly inhibited the proliferation, colony formation, invasion and migration, but induced the apoptosis of HCC cells. In line with this, downregulation of RP11-295G20.2 in HCC lines markedly suppressed the tumor growth in vivo. Mechanistically, RP11-295G20.2 could upregulate CCNB1 through targeting miR-6884-3p. More importantly, our rescue experiments revealed that miR-6884-3p/CCNB1 axis was involved in RP11-295G20.2-meditated tumorigenic behaviors of HCC cells.Conclusions: RP11-295G20.2 can contribute to HCC progression at least partly via the miR-6884-3p/CCNB1 axis, suggesting that RP11-295G20.2 may be a potential target for HCC therapy.Methods: RT-qPCR was employed to examine the expression levels of RP11-295G20.2, miR-6884-3p, and CCNB1 in HCC tissues and cell lines. CCK8 assay, transwell assay, colony formation assay and flow cytometry analysis were performed to evaluate the biological function of RP11-295G20.2 in HCC cells. The xenograft tumor assay was used to assess the effect of RP11-295G20.2 on the in vivo growth of HCC cells. The luciferase reporter assay, RIP assay and Spearman's correlation analysis were performed to explore the potential mechanisms underlying the roles of RP11-295G20.2 in HCC progression.
- Research Article
34
- 10.1016/j.canlet.2017.11.003
- Nov 8, 2017
- Cancer Letters
AGBL2 promotes cancer cell growth through IRGM-regulated autophagy and enhanced Aurora A activity in hepatocellular carcinoma
- Research Article
84
- 10.1016/j.celrep.2022.111194
- Aug 1, 2022
- Cell Reports
S-palmitoylation of PCSK9 induces sorafenib resistance in liver cancer by activating the PI3K/AKT pathway.
- Research Article
114
- 10.3892/ijo.2015.3309
- Dec 28, 2015
- International Journal of Oncology
MicroRNAs (miRNAs) have been found to play fundamental roles in the pathogenesis of hepatocellular carcinoma (HCC). Previous miRNA array data showed that miR-519a was upregulated in HCC tissues compared to adjacent non-tumor tissues. However, the functional role of miR-519a in HCC remains unexplored. In this study, we demonstrated that the expression of miR-519a was elevated in both HCC tissues and cell lines. Clinical association analysis revealed that high expression of miR-519a was correlated with adverse clinicopathological characteristics including large tumor size, high Edmondson-Steiner grading, advanced tumor-node-metastasis (TNM) tumor stage and venous infiltration. Furthermore, high expression of miR-519a conferred a reduced 5-year overall survival and disease-free survival of HCC patients. Moreover, we disclosed that miR-519a overexpression promoted, but miR-519a silencing reduced, HCC cell proliferation and cell cycle progression in vitro. Notably, we identified phosphatase and tensin homolog (PTEN) as a direct downstream target and functional mediator of miR-519a in HCC cells. Mechanistically, phosphatidylinositol-3-OH kinase (PI3K)/AKT pathway, downstream of PTEN, is essential for the functional roles of miR-519a in HCC cells. In conclusion, our results indicate that miR-519a promotes tumor growth of HCC by targeting PTEN-mediated PI3K/AKT pathway, and potentially serves as a novel prognostic biomarker and therapeutic target for HCC.
- Research Article
- 10.18282/amor.v2.i4.157
- Aug 30, 2016
- Advances in Modern Oncology Research
Cofactor of BRCA1: A new genetic marker for common malignant liver cancer
- Research Article
82
- 10.1053/j.gastro.2007.08.038
- Nov 1, 2007
- Gastroenterology
The biological aggressiveness of hepatocellular carcinoma (HCC) and the lack of optimal therapeutic strategies have rendered the disease a major challenge. Highly heterogeneous genetic alteration profiles of HCC have made it difficult to identify effective tailor-made molecular therapeutic targets. Therefore, classification of HCC into genetically homogeneous subclasses would be of great worth to develop novel therapeutic strategies. We clarified genome-scale chromosomal copy number alteration profiles and mutational statuses of p53 and beta-catenin in 87 HCC tumors. We investigated the possibility that HCC might be classifiable into a number of homogeneous subclasses based solely on their genetic alteration profiles. We also explored putative molecular therapeutic targets specific for each HCC subgroup. Unsupervised hierarchical cluster analysis based on chromosomal alteration profiles suggested that HCCs with heterogeneous genetic backgrounds are divisible into homogeneous subclasses that are highly associated with a range of clinicopathologic features of the tumors and moreover with clinical outcomes of the patients (P < .05). These genetically homogeneous subclasses could be characterized distinctively by pathognomonic chromosomal amplifications (eg, c-Myc-induced HCC, 6p/1q-amplified HCC, and 17q-amplified HCC). An in vitro experiment raised a possibility that Rapamycin would significantly inhibit the proliferative activities of HCCs with 17q amplification. HCC is composed of several genetically homogeneous subclasses, each of which harbors characteristic genetic alterations that can be putative tailor-made molecular therapeutic targets for HCCs with specific genetic backgrounds. Our results offer an opportunity for developing novel individualized therapeutic modalities for distinctive genome types of HCC.
- Research Article
7
- 10.3389/fonc.2020.607593
- Jan 8, 2021
- Frontiers in Oncology
Hepatocellular carcinoma (HCC) is a common malignant liver tumor worldwide. Tumor recurrence and metastasis contribute to the bad clinical outcome of HCC patients. Substantial studies have displayed lncRNAs modulate various tumorigenic processes of many cancers. Our current work was aimed to investigate the function of LINC00675 in HCC and to recognize the potential interactions between lncRNAs and microRNAs. GFI1 can exhibit a significant role in the progression of human malignant tumors. Firstly, GFI1 was identified using real-time PCR in HCC tissues and cells. In this work, we indicated GFI1 was remarkably reduced in HCC tissues and cells. Meanwhile, GFI1 specifically interacted with the promoter of LINC00675. Up-regulation of LINC00675 obviously repressed the migration and invasion capacity of SMCC-7721 and QGY-7703 cells in vitro. Moreover, decrease of LINC00675 competitively bound to miR-942-5p that contributed to the miRNA-mediated degradation of GFI1, thus facilitated HCC metastasis. The ceRNA function of LINC00675 in HCC cells was assessed and confirmed using RNA immunoprecipitation assay and RNA pull-down assays in our work. Additionally, we proved overexpression of miR-942-5p promoted HCC progression, which was reversed by the up-regulation of GFI1. In summary, LINC00675 might act as a prognostic marker for HCC, which can inhibit HCC development via regulating miR-942-5p and GFI1.
- Research Article
10
- 10.14218/jcth.2021.00111
- Jul 23, 2021
- Journal of Clinical and Translational Hepatology
Background and AimsAT-rich interactive domain-containing protein 1A (ARID1A) is frequently mutated or deficient in hepatocellular carcinoma (HCC). However, the role of ARID1A in HCC remains unclear. Therefore, the biological role of ARID1A in HCC was evaluated and a potential mechanism was investigated.MethodsArid1a was knocked out in the livers of mice using the CRISPR/Cas9 system delivered by hydrodynamic tail vein injection. The development of HCC was observed in different mouse models. The correlation of ARID1A and prognosis in patients with HCC was analyzed using cBioPortal. The effect of ARID1A on cell proliferation was assessed by MTT assay following the manipulation of candidate genes.ResultsARID1A deficiency alone did not cause HCC in mice, but knockout of ARID1A accelerated liver tumorigenesis in response to diethylnitrosamine (DEN) or when a combination knockout of phosphatase and tensin homolog (Pten) plus tumor protein P53 (p53) was introduced. ARID1A mutations were associated with a poorer prognosis in HCC patients. The mRNA level of MYC was significantly higher in patients with an ARID1A mutation compared to those without a mutation. Ectopic expression of ARID1A inhibited HCC cell proliferation. ARID1A knockout increased HCC cell growth and resulted in disruptions to DNA damage repair and apoptosis following radiation stress. Furthermore, mechanistic studies revealed that ARID1A inhibited the proliferation of HCC cells via transcriptional down-regulation of MYC.ConclusionsThese results describe ARID1A as a tumor suppressor in the liver. A deficiency in ARID1A predicts worse survival in HCC patients and promotes HCC progression via up-regulation of MYC transcription.
- Research Article
14
- 10.1002/mc.23794
- Jul 17, 2024
- Molecular carcinogenesis
Hepatocellular carcinoma (HCC) is a common malignant tumor. Histone lactylation, a novel epigenetic modification, plays a crucial role in various cancers. However, the functional role and underlying mechanism of histone lactylation in HCC progression have not yet been investigated. Histone lactylation levels in HCC tissues and cells were assessed using a densitometric kit and western blot analysis. The role of histone lactylation in cell malignant phenotypes was determined through functional assays in vitro, and a xenograft tumor model was established to verify the function of histone lactylation in vivo. ChIP assay was performed to explore the interaction between histone lactylation and endothelial cell-specific molecule 1 (ESM1). Additionally, gain-and-loss-of-function assays were conducted to investigate the regulatory role of ESM1 in HCC pathogenesis. Histone lactylation levels were increased in HCC tissues and cells, and H3K9 lactylation (H3K9la) and H3K56 lactylation (H3K56la) were identified as the histone modification sites. We observed that H3K9la and H3K56la caused abnormal histone lactylation and were associated with poor prognosis. Functionally, histone lactylation was found to promote HCC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) process in vitro. However, histone lactylation inhibition with 2-deoxy-d-glucose (2-DG) reduced the malignant phenotypes of HCC cells. In vivo, 2-DG treatment reduced tumor growth and metastasis in the HCC mouse model. Mechanistically, it was revealed that histone lactylation activated ESM1 transcription in HCC cells. ESM1 was expressed at a high level in HCC and exerted a carcinogenic role. Histone lactylation facilitates cell malignant phenotypes, tumor growth, and metastasis by upregulating ESM1 expression in HCC, which reveals the downstream molecular mechanism of histone lactylation and might provide a novel therapeutic target for HCC therapy.
- Research Article
22
- 10.2147/ott.s269606
- Oct 1, 2020
- OncoTargets and Therapy
BackgroundThe ubiquitin-proteasome system participates in the pathogenesis and progression of hepatocellular carcinoma (HCC). As an E3 ubiquitin ligase, RNF128 has been proved vital in carcinogenesis, whereas, little is known about the oncogenic mechanisms of RNF128 in HCC.Materials and MethodsThrough tissue microarray from HCC patients, we analyzed RNF128 expression and its relationship with clinical outcomes in HCC. Western blot and quantitative realtime polymerase chain reaction (qRT-PCR) were performed to examine expression levels of RNF128 in HCC tissues and cell lines. Effects of RNF128 on HCC cellular biological functions and the potential mechanism were evaluated through knockdown and overexpression assays in vitro and in vivo methods.ResultsRNF128 expression was found to be remarkably elevated in HCC tissues compared with adjacent normal tissues. Furthermore, the overexpression of RNF128 enhanced hepatoma cells proliferation, colony formation, migration, invasion, and apoptotic resistance both in vitro and in vivo. Mechanistically, RNF128 activated EGFR/MEK/ERK signaling pathway and the EGFR inhibitor, gefitinib partially reversed RNF128-enhanced proliferation, invasion, and migration in hepatoma cells.ConclusionRNF128 promotes HCC progression by activating EGFR/MEK/ERK signaling pathway, which might function as a novel prognostic molecular signature with the potential to be a candidate therapeutic target for HCC patients.
- Front Matter
4
- 10.1053/j.gastro.2008.01.055
- Mar 1, 2008
- Gastroenterology
Rho GTPase and Wnt Signaling Pathways in Hepatocarcinogenesis
- New
- Research Article
- 10.1016/j.cmet.2025.10.007
- Nov 1, 2025
- Cell metabolism
- New
- Addendum
- 10.1016/j.cmet.2025.10.019
- Nov 1, 2025
- Cell metabolism
- New
- Addendum
- 10.1016/j.cmet.2025.10.023
- Nov 1, 2025
- Cell metabolism
- New
- Research Article
- 10.1016/j.cmet.2025.10.008
- Nov 1, 2025
- Cell metabolism
- New
- Addendum
- 10.1016/j.cmet.2025.10.020
- Nov 1, 2025
- Cell metabolism
- New
- Research Article
- 10.1016/j.cmet.2025.10.002
- Nov 1, 2025
- Cell metabolism
- New
- Addendum
- 10.1016/j.cmet.2025.10.024
- Nov 1, 2025
- Cell metabolism
- New
- Research Article
- 10.1016/j.cmet.2025.10.004
- Nov 1, 2025
- Cell metabolism
- New
- Research Article
- 10.1016/j.cmet.2025.10.003
- Oct 31, 2025
- Cell metabolism
- Research Article
- 10.1016/j.cmet.2025.10.001
- Oct 24, 2025
- Cell metabolism
- Ask R Discovery
- Chat PDF
AI summaries and top papers from 250M+ research sources.