1.DNA Methylation and Uveal Melanoma.
Zhi-Kun YANG ; Jing-Yun YANG ; ; Zhuo-Zai XU ; Wei-Hong YU
Chinese Medical Journal 2018;131(7):845-851
ObjectiveThe objective of the study was to summarize the role of DNA methylation in the development and metastasis of uveal melanoma (UM).
Data SourcesThe relevant studies in MEDLINE were searched.
Study SelectionIn this review, we performed a comprehensive literature search in MEDLINE using "uveal melanoma" AND ("DNA methylation" OR "epigenetics") for original research/review articles published before February 2018 on the relationship between DNA methylation and UM. References of the retrieved studies were also examined to search for potentially relevant papers.
ResultsPrevious studies on the relationship between DNA methylation and UM covered many genes including tumor suppressor genes (TSGs), cyclin-dependent kinase genes, and other genes. Among them, the TSG genes such as RASSF1A and p16INK4a, which encodes a cyclin-dependent kinase inhibitor, are relatively well-studied genes. Specifically, a high percentage of promoter methylation of RASSF1A was observed in UM cell lines and/or patients with UM. Promoter methylation of RASSF1A was also associated with the development of metastasis. Similarly, a high percentage of promoter hypermethylation of p16INK4a was found in UM cell lines. DNA promoter methylation can control the expression of p16INK4a, which affect cell growth, migration, and invasion in UM. Many other genes might also be involved in the pathogenesis of UM such as the Ras and EF-hand domain containing (RASEF) gene, RAB31, hTERT, embryonal fyn-associated substrate, and deleted in split-hand/split-foot 1.
ConclusionsOur review reveals the complex mechanisms underlying the tumorigenesis of UM and highlights the great needs of future studies to discover more genes/5'-C-phosphate-G-3' sites contributing to the development/metastasis of UM and explore the mechanisms through which epigenetic changes exert their function in UM.
Cell Transformation, Neoplastic ; genetics ; DNA Methylation ; genetics ; Epigenesis, Genetic ; genetics ; Humans ; Melanoma ; genetics ; Promoter Regions, Genetic ; genetics ; Uveal Neoplasms ; genetics
2.Palmitoylation of GNAQ/11 is critical for tumor cell proliferation and survival in GNAQ/11-mutant uveal melanoma.
Yan ZHANG ; Baoyuan ZHANG ; Yongyun LI ; Yuting DAI ; Jiaoyang LI ; Donghe LI ; Zhizhou XIA ; Jianming ZHANG ; Ping LIU ; Ming CHEN ; Bo JIAO ; Ruibao REN
Frontiers of Medicine 2022;16(5):784-798
More than 85% of patients with uveal melanoma (UM) carry a GNAQ or GNA11 mutation at a hotspot codon (Q209) that encodes G protein α subunit q/11 polypeptides (Gαq/11). GNAQ/11 relies on palmitoylation for membrane association and signal transduction. Despite the palmitoylation of GNAQ/11 was discovered long before, its implication in UM remains unclear. Here, results of palmitoylation-targeted mutagenesis and chemical interference approaches revealed that the loss of GNAQ/11 palmitoylation substantially affected tumor cell proliferation and survival in UM cells. Palmitoylation inhibition through the mutation of palmitoylation sites suppressed GNAQ/11Q209L-induced malignant transformation in NIH3T3 cells. Importantly, the palmitoylation-deficient oncogenic GNAQ/11 failed to rescue the cell death initiated by the knock down of endogenous GNAQ/11 oncogenes in UM cells, which are much more dependent on Gαq/11 signaling for cell survival and proliferation than other melanoma cells without GNAQ/11 mutations. Furthermore, the palmitoylation inhibitor, 2-bromopalmitate, also specifically disrupted Gαq/11 downstream signaling by interfering with the MAPK pathway and BCL2 survival pathway in GNAQ/11-mutant UM cells and showed a notable synergistic effect when applied in combination with the BCL2 inhibitor, ABT-199, in vitro. The findings validate that GNAQ/11 palmitoylation plays a critical role in UM and may serve as a promising therapeutic target for GNAQ/11-driven UM.
Humans
;
Mice
;
Animals
;
Lipoylation
;
NIH 3T3 Cells
;
Uveal Neoplasms/genetics*
;
Melanoma/genetics*
;
Cell Proliferation
;
Proto-Oncogene Proteins c-bcl-2
;
GTP-Binding Protein alpha Subunits, Gq-G11/genetics*
3.MicroRNA 145 may play an important role in uveal melanoma cell growth by potentially targeting insulin receptor substrate-1.
Yang LI ; Qiming HUANG ; Xuehui SHI ; Xiang JIN ; Li SHEN ; Xiaolin XU ; Wenbin WEI ;
Chinese Medical Journal 2014;127(8):1410-1416
BACKGROUNDMicroRNAs (miRNAs) contribute to tumorigenesis by acting as either oncogenes or tumor suppressor genes. In this study, we investigated the role of miR-145 in the pathogenesis of uveal melanoma.
METHODSExpression profiles of miRNAs in uveal melanoma were performed using Agilent miRNA array. Quantitative real-time polymerase chain reaction was used to screen the expression levels of miR-145 in normal uveal tissue, uveal melanoma tissue, and uveal melanoma cell lines. Lenti-virus expression system was used to construct MUM-2B and OCM-1 cell lines with stable overexpression of miR-145. Cell proliferation, cell cycle, and cell apoptosis of these miR-145 overexpression cell lines were examined by MTT assay and flow cytometry respectively. The target genes of miR-145 were predicted by bioinformatics and confirmed using a luciferase reporter assay. The expression of insulin-like growth factor-1 receptor (IGF-1R), insulin receptor substrate-1 (IRS-1) proteins was determined by Western blotting analysis. IRS-1 was knocked down in OCM-1 cells. TUNEL, BrdU, and flow cytometry assay were performed in IRS-1 knocked down OCM-1 cell lines to analyze its function.
RESULTSForty-seven miRNAs were up regulated in uveal melanoma and 61 were down regulated. miR-145 expression was significantly lower in uveal melanoma sample and the cell lines were compared with normal uveal sample. Overexpression of miR-145 suppressed cell proliferation by blocking the G1 phase entering S phase in uveal melanoma cells, and promoted uveal melanoma cell apoptosis. IRS-1 was identified as a potential target of miR-145 by dual luciferase reporter assay. Knocking down of IRS-1 had similar effect as overexpression of miR-145.
CONCLUSIONmiR-145 might act as a tumor suppressor in uveal melanoma, and downregulation of the target IRS-1 might be a potential mechanism.
Apoptosis ; genetics ; physiology ; Blotting, Western ; Cell Cycle ; genetics ; physiology ; Cell Line, Tumor ; Cell Proliferation ; genetics ; physiology ; Humans ; In Vitro Techniques ; Insulin Receptor Substrate Proteins ; genetics ; metabolism ; Melanoma ; genetics ; metabolism ; pathology ; MicroRNAs ; genetics ; metabolism ; Polymerase Chain Reaction ; Uveal Neoplasms ; genetics ; metabolism ; pathology