Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 83(21): 3544-3561, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37352379

ABSTRACT

The tumor microenvironment is distinctive in primary and secondary liver cancer. B cells represent an important component of immune infiltrates. Here, we demonstrated that B cells are an important regulator in hepatocellular carcinoma (HCC) and colorectal cancer liver metastasis (CRLM) microenvironments. B cells displayed distinct developmental trajectories in HCC and CRLM. Single-cell analysis revealed that IgG+ plasma cells preferentially accumulated in HCC, whereas IgA+ plasma cells were preferentially enriched in CRLM. Mechanistically, IgG+ plasma cells in HCC were recruited by tumor-associated macrophages via the CXCR3-CXCL10 axis, whereas IgA+ plasma cells in CRLM were recruited by metastatic tumor cells via CCR10-CCL28 signaling. Functionally, IgG+ plasma cells preferentially promoted protumorigenic macrophages formation in HCC, and IgA+ plasma cells preferentially induced granulocytic myeloid-derived suppressor cells activation in CRLM. Clinically, increased infiltration of IgG+ plasma cells and macrophages in HCC was correlated to worse survival, whereas increased intratumoral IgA+ plasma cells and neutrophils in CRLM indicated poor prognosis. Taken together, this study demonstrated plasma and myeloid cell-mediated immunosuppression in HCC and CRLM, suggesting that selectively modulating primary or secondary tumor-related immunosuppressive regulatory networks might reprogram the microenvironment and provide an immunotherapeutic strategy for treating liver cancer. SIGNIFICANCE: The immunomodulatory patterns of tumor-infiltrating B cells are distinct in primary and secondary liver cancer, with plasma cells mediating important physiologic processes that drive cancer progression.


Subject(s)
Carcinoma, Hepatocellular , Colorectal Neoplasms , Liver Neoplasms , Humans , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/pathology , Signal Transduction , Colorectal Neoplasms/pathology , Immunoglobulin G , Immunoglobulin A , Tumor Microenvironment
2.
Cancer Res ; 83(1): 89-102, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36102722

ABSTRACT

The tyrosine kinase inhibitor lenvatinib is a first-line drug for treating patients with advanced hepatocellular carcinoma (HCC). However, its efficacy is severely hampered by drug resistance. Insights into the molecular mechanisms underlying lenvatinib resistance could provide new strategies to improve and prolong responses. Here, we performed unbiased proteomic screening of parental and lenvatinib-resistant HCC cells and discovered that methyltransferase-like protein-1 (METTL1) and WD repeat domain 4 protein (WDR4), the two key components of the tRNA N7-methylguanosine (m7G) methyltransferase complex, were dramatically upregulated in lenvatinib-resistant cells. METTL1 knockdown overrode resistance by impairing the proliferation capacity of HCC cells and promoting apoptosis under lenvatinib treatment. In addition, overexpression of wild-type METTL1 but not its catalytic dead mutant induced lenvatinib resistance. Animal experiments including hydrodynamic injection, subcutaneous implantation, and orthotopic xenograft mouse models further demonstrated the critical function of METTL1/WDR4-mediated m7G tRNA modification in promoting lenvatinib resistance in vivo. Mechanistically, METTL1 promoted translation of EGFR pathway genes to trigger drug resistance. This work reveals the important role of METTL1-mediated m7G tRNA modification in promoting lenvatinib resistance and provides a promising prediction marker and intervention target for resistance. SIGNIFICANCE: Upregulation of tRNA m7G methyltransferase complex components METTL1 and WDR4 promotes lenvatinib resistance in HCC and confers a sensitivity to METTL1 targeting, providing a promising strategy to override resistance.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Proteomics , RNA, Transfer/genetics , GTP-Binding Proteins/metabolism
3.
Int J Gen Med ; 15: 1891-1899, 2022.
Article in English | MEDLINE | ID: mdl-35221718

ABSTRACT

PURPOSE: miR-500a-3p has been extensively reported to be implicated in the development and progression in several human cancer types. This study aimed to investigate the diagnostic and prognostic significance of miR-500a-3p as a biomarker in hepatocellular carcinoma (HCC). METHODS: miR-500a-3p expression was evaluated by in situ hybridization (ISH) and real-time PCR in 10 adjacent normal tissues (ANT), 21 liver fibrosis tissues, and 110 HCC tissues. Statistical analysis was used to investigate the correlation of miR-500a-3p expression with clinicopathological features in HCC patients. Kaplan-Meier survival analysis was performed to evaluate the prognostic significance of miR-500a-3p in overall survival and recurrence-free survival in HCC patients. RESULTS: In this study, we found that expression levels of miR-500a-3p were enhanced in HCC tissues. High miR-500a-3p levels were positively correlated with multiple clinicopathological features, including advanced clinical stage, distant metastatic status, increased AFP levels and poor tumor differentiation degree. More importantly, high miR-500a-3p levels predicted poor overall survival and early recurrence in HCC patients. Finally, a strong and positive correlation of miR-500a-3p mRNA expression with ISH staining scores was observed in clinical HCC tissues. CONCLUSION: Our findings suggest that miR-500a-3p might be used as a novel biomarker to facilitate early diagnosis and predict prognosis in HCC patients.

4.
Anticancer Agents Med Chem ; 22(2): 254-260, 2022.
Article in English | MEDLINE | ID: mdl-33797381

ABSTRACT

BACKGROUND: SphK1 is a conserved lipid kinase, which can catalyze the formation of tumorpromoting factor sphingosine phosphate-1 (S1P). OBJECTIVE: This study aimed to investigate the effect of SphK1 on the proliferation/migration of colon cancer cells and associated mechanisms. METHODS: Transcription of the SphK1 gene in colon cancer cells was detected. Gene transcription of SphK1 was inhibited by transfecting with the si-SphK1 gene in colon cancer cells. Effects of SphK1 inhibition (si-SphK1) on cell migration/proliferation were detected using the transwell system and MTS. Gene transcription of SIP, S1PR1, S1PR2, S1PR3, and activation of JAK/STAT3 pathway were examined using RT-PCR and western blot assay. S1PR1 over-expressing plasmid was constructed and transfected into cells. Effects of S1PR1 overexpression on migration/proliferation of si-SphK1 transfected colon cancer cells and activation of JAK/STAT3 pathway were determined using RT-PCR and western blotting. RESULTS: Gene transcription of SphK1 in SW480 and HT-29 colon cancer cells was significantly inhibited by transfection of the si-SphK1 gene. Transwell migration and MTS findings showed that si-SphK1 transfection (si- SphK1 group) could reduce migration quantity and cell viability of colon cancer cells compared to the negative control (NC) (p<0.0001). SphK1 inhibition (si-SphK1 group) significantly down-regulated S1PR1 and S1PR3 gene transcription in SW480 and HT-29 cells (p<0.0001) and decreased activation level of JAKSTAT3 signaling pathway compared to NC group (p<0.05). Over-expression of S1PR1 reversed inhibitory effects of si-SphK1 on migration/proliferation of SW480 and activation of JAK/Stat3. CONCLUSION: SphK1 promoted proliferation and migration of colon cancer cells through promoting JAK/STAT activation and up-regulating S1PR1 expression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/metabolism , Janus Kinase 1/metabolism , STAT3 Transcription Factor/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Up-Regulation , Adaptor Proteins, Signal Transducing/genetics , Cell Movement , Cell Proliferation , Colonic Neoplasms/pathology , Humans , Sphingosine-1-Phosphate Receptors/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...