Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Chem Biol Interact ; 387: 110809, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38006958

ABSTRACT

BACKGROUND: Hydroquinone (HQ), a major metabolite of benzene and known hematotoxic carcinogen. MicroRNA 1246 (miR-1246), an oncogene, regulates target genes in carcinogenesis including leukemia. This study investigates the impact of exosomal derived miR-1246 from HQ-transformed (HQ19) cells on cell-to-cell communication in recipient TK6 cells. METHODS: RNA sequencing was used to identify differentially expressed exosomal miRNAs in HQ19 cells and its phosphate buffered solution control cells (PBS19), which were then confirmed using qRT-PCR. The impact of exosomal miR-1246 derived from HQ-transformed cells on cell cycle distribution was investigated in recipient TK6 cells. RESULTS: RNA sequencing analysis revealed that 34 exosomal miRNAs were upregulated and 158 miRNAs were downregulated in HQ19 cells compared with PBS19 cells. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses predicted that their targets are enriched in cancer development-related pathways, such as MAPK signaling, microRNAs in cancer, apoptosis, PI3K-Akt signaling, cell cycle, Ras signaling, and Chronic myeloid leukemia. Eleven miRNAs were confirmed to have differential expression through qRT-PCR, with 6 upregulated (miR-140-3p, miR-551b-3p, miR-7-5p, miR-1290, miR-92a-3p, and miR-1246) and 5 downregulated (miR-183-5p, miR-26a-5p, miR-30c-5p, miR-205-5p, and miR-99b-3p). Among these, miR-1246 exhibited the highest expression level. HQ exposure resulted in a concentration-dependent increase in miR-1246 levels and decrease Cyclin G2 (CCNG2) levels in TK6 cells. Similarly, exosomes from HQ19 exhibited similar effects as HQ exposure. Dual luciferase reporter gene assays indicated that miR-1246 could band to CCNG2. After HQ exposure, exosomal miR-1246 induced cell cycle arrest at the S phase, elevating the expression of genes like pRb, E2F1, and Cyclin D1 associated with S phase checkpoint. However, silencing miR-1246 caused G2/M-phase arrest. CONCLUSION: HQ-transformed cells' exosomal miR-1246 targets CCNG2, regulating TK6 cell cycle arrest, highlighting its potential as a biomarker for HQ-induced malignant transformation.


Subject(s)
Cyclin G2 , MicroRNAs , Humans , Cyclin G2/genetics , Cyclin G2/metabolism , S Phase , Hydroquinones/toxicity , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Transformation, Neoplastic
2.
BMC Cancer ; 21(1): 1074, 2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34598688

ABSTRACT

BACKGROUND: The human miR-17-92 polycistron is the first reported and most well-studied onco-miRNA with a cluster of seven miRNAs. miR-17-5p, a member of the miR-17-92 family, plays an important role in tumor cell proliferation, apoptosis, migration and invasion. However, few studies have shown the role of miR-17-5p in the cell cycle of head and neck squamous cell carcinoma (HNSCC). METHODS: RT-qPCR was used to detect miR-17-5p expression levels in 64 HNSCC tissues and 5 cell lines. The relationship between the expression of miR-17-5p in the tissues and the clinical characteristics of the patients was analyzed. HNSCC cells were transfected with an miR-17-5p mimic or inhibitor to evaluate cell cycle distribution by flow cytometry. Cell cycle distribution of cells transfected with target gene was evaluated using flow cytometry. Dual-luciferase reporter assay was used to detect the regulatory effect of miR-17-5p on target gene expression. RESULTS: In the present study, we found that miR-17-5p expression in HNSCC tissues and cell lines was remarkably increased, and miR-17-5p is related to recurrence in HNSCC patients. Silencing miR-17-5p blocked HNSCC cells in G2/M phase, whereas its overexpression propelled cell cycle progression. More importantly, we verified that miR-17-5p negatively regulated CCNG2 mRNA and protein expression by directly targeting its 3'UTR. CONCLUSION: These findings suggest that miR-17-5p might act as a tumor promoter and prognostic factor for recurrence in HNSCC patients.


Subject(s)
Cyclin G2/metabolism , G2 Phase Cell Cycle Checkpoints , Head and Neck Neoplasms/metabolism , M Phase Cell Cycle Checkpoints , MicroRNAs/metabolism , Neoplasm Recurrence, Local/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , 3' Untranslated Regions/genetics , Apoptosis/genetics , Area Under Curve , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin G2/genetics , Down-Regulation , Female , Gene Silencing , Head and Neck Neoplasms/genetics , Humans , Luciferases/metabolism , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Recurrence, Local/metabolism , RNA, Messenger/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Transfection , Up-Regulation
3.
FASEB J ; 35(2): e21169, 2021 02.
Article in English | MEDLINE | ID: mdl-33205477

ABSTRACT

Disruption of extravillous trophoblast (EVT) migration and invasion is considered to be responsible for pathological placentation in preeclampsia (PE). Cyclin G2 (CCNG2) is an atypical cyclin that inhibits cell cycle progression. However, its biological function and underlying molecular mechanism in PE are poorly understood. In this study, clinical data demonstrated that CCNG2 was significantly upregulated in PE placenta and associated with invasive EVT dysfunction. Additionally, Ccng2 knockout led to an attenuation of PE-like symptoms in the PE mouse model produced via treatment with NG-nitro-L-arginine methyl ester (L-NAME). In vitro, CCNG2 inhibited the migration, invasion, and endothelial-like network formation of human trophoblast cell line HTR8/SVneo. Mechanically, CCNG2 suppressed JNK-dependent Wnt/PCP signaling and its downstream indicators including epithelial-to-mesenchymal transition (EMT) markers and matrix metalloproteinases (MMPs) via promoting the polyubiquitination degradation of dishevelled 2 (Dvl2) protein in HTR8/SVneo cells. We also discovered that the E3 ligase Ring finger protein 123 (RNF123), as a novel CCNG2 target among HTR8/SVneo cells, interacted with Dvl2 and participated in CCNG2-induced polyubiquitination degradation of Dvl2. Moreover, we verified that the treatment of HTR8/SVneo cells with RNF123-specific siRNA improved polyubiquitination-induced degradation of Dvl2 and the activity of Wnt/PCP-JNK signaling mediated by CCNG2. Taken together, our results reveal that the CCNG2/RNF123/Dvl2/JNK axis may be involved in the pathogenesis and progression of PE through trophoblastic cell function modulation, thus probably providing us with new therapeutic strategies for PE treatment.


Subject(s)
Cell Movement/genetics , Cyclin G1/metabolism , Cyclin G2/metabolism , Dishevelled Proteins/metabolism , MAP Kinase Signaling System/genetics , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , Ubiquitin-Protein Ligases/metabolism , Up-Regulation/genetics , Adult , Animals , Cell Line , Cyclin G1/genetics , Cyclin G2/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Pregnancy , Transfection , Ubiquitin-Protein Ligases/genetics
4.
Life Sci ; 265: 118858, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33290791

ABSTRACT

AIMS: Previous studies have shown that the forkhead transcription factor FoxO6 involved in memory consolidation and hepatic glucose homeostasis. Here we asked whether chicken FoxO6 may regulate preadipocyte proliferation, apoptosis and early adipogenesis. MAIN METHODS: Overexpression and knockdown of FoxO6 were performed and evaluated through cell proliferation methods, Oil-Red-O staining, and specific marker expression. Chromatin immunoprecipitation (ChIP) assay was performed to confirm cyclin G2 (CCNG2) as a direct target gene of FoxO6. KEY FINDINGS: FoxO6 is ubiquitously expressed in different chicken tissues and highly expressed in liver, abdominal fat, and preadipocytes in cultured cell. FoxO6 overexpression decreased preadipocyte proliferation by causing G1-phase cell-cycle arrest, whereas inhibition of FoxO6 showed the opposite effects. Overexpression or knockdown of FoxO6 significantly altered the mRNA and protein levels of cell-cycle related markers, such as CCNG2, cyclin dependent kinase inhibitor 1B (CDKN1B), cyclin dependent kinase inhibitor 1A (CDKN1A) and cyclin D2 (CCND2). During preadipocyte proliferation, FoxO6 targets and induces expression of CCNG2, as confirmed by ChIP assay and qPCR. In addition, FoxO6 induces preadipocyte apoptosis through increasing the protein expression levels of cleaved caspase-3 and cleaved caspase-8. Moreover, FoxO6 at the early stage of adipogenesis suppressed mRNA and protein levels of the key early regulators of adipogenesis, such as PPARγ and C/EBPα. SIGNIFICANCE: The results demonstrate that FoxO6 controls preadipocyte proliferation, apoptosis and early adipogenesis, and point to new approaches for further studies related to obesity.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Apoptosis/genetics , Cell Proliferation/genetics , Forkhead Transcription Factors/genetics , Adipocytes/cytology , Animals , Cells, Cultured , Chickens , Chromatin Immunoprecipitation , Cyclin G2/genetics , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques , Male , PPAR gamma/metabolism
5.
IUBMB Life ; 72(7): 1491-1503, 2020 07.
Article in English | MEDLINE | ID: mdl-32196930

ABSTRACT

Small extracellular vesicle (sEV) has precise impacts on tumor microenvironment and play vital functions in intercellular interaction. However, the functional role of sEV miRNA on laryngeal squamous cell carcinoma (LSCC) is largely unresolved. Here, the expression of miR-1246 in LSCC tissues and plasma sEV was examined. The internalization ability of sEV was determined by uptake assay. Then, the source and purity of sEV were checked through RNase and/or pharmacological inhibitors application. The invasion, migration, proliferation, and cell cycle assays were used to determine the altered abilities of miR-1246 in sEV in LSCC. Finally, target gene of miR-1246, Cyclin G2 (CCNG2), was stained immunohistochemically. In addition, the relationship between CCNG2 and clinicopathological features of patients was analyzed. We found that miR-1246 was higher in LSCC tissues and plasma sEV. MiR-1246 was enriched in sEV rather than soluble form. SEV could be internalized into adjacent cells. Lack of miR-1246 in sEV abrogated the tumorigenesis of LSCC. Furthermore, CCNG2 knockdown arrested the cell cycle and correlated to clinicopathological features and prognosis of LSCC patients. Taken together, we found that the function of sEV miR-1246 by regulating CCNG2 is responsible for LSCC advancement with emphasis on the main source of miR-1246 mainly root in sEV rather than in soluble form.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/pathology , Carcinoma, Squamous Cell/pathology , Cyclin G2/metabolism , Gene Expression Regulation, Neoplastic , Laryngeal Neoplasms/pathology , MicroRNAs/genetics , Aged , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Cyclin G2/genetics , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/metabolism , Neoplasm Invasiveness , Prognosis , Survival Rate , Tumor Cells, Cultured
6.
Int J Mol Sci ; 20(8)2019 Apr 12.
Article in English | MEDLINE | ID: mdl-31013711

ABSTRACT

Ovarian cancer is the leading cause of death from gynecological cancers. MicroRNAs (miRNAs) are small, non-coding RNAs that interact with the 3' untranslated region (3' UTR) of target genes to repress their expression. We have previously reported that miR-590-3p promoted ovarian cancer growth and metastasis, in part by targeting Forkhead box A (FOXA2). In this study, we further investigated the mechanisms by which miR-590-3p promotes ovarian cancer development. Using luciferase reporter assays, real-time PCR, and Western blot analyses, we demonstrated that miR-590-3p targets cyclin G2 (CCNG2) and Forkhead box class O3 (FOXO3) at their 3' UTRs. Silencing of CCNG2 or FOXO3 mimicked, while the overexpression of CCNG2 or FOXO3 reversed, the stimulatory effect of miR-590-3p on cell proliferation and invasion. In hanging drop cultures, the overexpression of mir-590 or the transient transfection of miR-590-3p mimics induced the formation of compact spheroids. Transfection of the CCNG2 or FOXO3 plasmid into the mir-590 cells resulted in the partial disruption of the compact spheroid formation. Since we have shown that CCNG2 suppressed ß-catenin signaling, we investigated if miR-590-3p regulated ß-catenin activity. In the TOPFlash luciferase reporter assays, mir-590 increased ß-catenin/TCF transcriptional activity and the nuclear accumulation of ß-catenin. Silencing of ß-catenin attenuated the effect of mir-590 on the compact spheroid formation. Taken together, these results suggest that miR-590-3p promotes ovarian cancer development, in part by directly targeting CCNG2 and FOXO3.


Subject(s)
Cyclin G2/genetics , Forkhead Box Protein O3/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Ovarian Neoplasms/genetics , RNA Interference , 3' Untranslated Regions , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Silencing , Genes, Reporter , Humans , Models, Biological , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured , Wnt Signaling Pathway
7.
Int J Biol Sci ; 15(3): 544-555, 2019.
Article in English | MEDLINE | ID: mdl-30745841

ABSTRACT

Cyclin G2 has been identified as a tumour suppressor in several cancers. However, its regulatory roles and underlying mechanisms in tumours are still unknown. In this study, we demonstrated that cyclin G2 was expressed at low levels in glioma, which was as a poor prognostic factor for this disease. We also found that, cyclin G2 could suppress cell proliferation, initiate cell apoptosis and reduce aerobic glycolysis, suggesting that cyclin G2 plays a tumour suppressive role in glioma. Mechanistically, cyclin G2 could negatively regulate tyrosine-10 phosphorylation of a critical glycolytic enzyme, lactate dehydrogenase A, through direct interaction. Taken together, these results indicate that cyclin G2 acts as a tumour suppressor in glioma by repressing glycolysis and tumour progression through its interaction with LDHA.


Subject(s)
Cell Proliferation/physiology , L-Lactate Dehydrogenase/metabolism , Wound Healing/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin G2/genetics , Cyclin G2/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Wound Healing/genetics
8.
J Exp Clin Cancer Res ; 37(1): 317, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30547803

ABSTRACT

BACKGROUND: Gastric cancer is one of the most common malignant tumors. Cyclin G2 has been shown to be associated with the development of multiple types of tumors, but its underlying mechanisms in gastric tumors is not well-understood. The aim of this study is to investigate the role and the underlying mechanisms of cyclin G2 on Wnt/ß-catenin signaling in gastric cancer. METHODS: Real-time PCR, immunohistochemistry and in silico assay were used to determine the expression of cyclin G2 in gastric cancer. TCGA datasets were used to evaluate the association between cyclin G2 expression and the prognostic landscape of gastric cancers. The effects of ectopic and endogenous cyclin G2 on the proliferation and migration of gastric cancer cells were assessed using the MTS assay, colony formation assay, cell cycle assay, wound healing assay and transwell assay. Moreover, a xenograft model and a metastasis model of nude mice was used to determine the influence of cyclin G2 on gastric tumor growth and migration in vivo. The effects of cyclin G2 expression on Wnt/ß-catenin signaling were explored using a TOPFlash luciferase reporter assay, and the molecular mechanisms involved were investigated using immunoblots assay, yeast two-hybrid screening, immunoprecipitation and Duolink in situ PLA. Ccng2-/- mice were generated to further confirm the inhibitory effect of cyclin G2 on Wnt/ß-catenin signaling in vivo. Furthermore, GSK-3ß inhibitors were utilized to explore the role of Wnt/ß-catenin signaling in the suppression effect of cyclin G2 on gastric cancer cell proliferation and migration. RESULTS: We found that cyclin G2 levels were decreased in gastric cancer tissues and were associated with tumor size, migration and poor differentiation status. Moreover, overexpression of cyclin G2 attenuated tumor growth and metastasis both in vitro and in vivo. Dpr1 was identified as a cyclin G2-interacting protein which was required for the cyclin G2-mediated inhibition of ß-catenin expression. Mechanically, cyclin G2 impacted the activity of CKI to phosphorylate Dpr1, which has been proved to be a protein that acts as a suppressor of Wnt/ß-catenin signaling when unphosphorylated. Furthermore, GSK-3ß inhibitors abolished the cyclin G2-induced suppression of cell proliferation and migration. CONCLUSIONS: This study demonstrates that cyclin G2 suppresses Wnt/ß-catenin signaling and inhibits gastric cancer cell growth and migration through Dapper1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cyclin G2/metabolism , Nuclear Proteins/metabolism , Stomach Neoplasms/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Chlorocebus aethiops , Cyclin G2/biosynthesis , Cyclin G2/genetics , Female , Genes, Tumor Suppressor , HT29 Cells , HeLa Cells , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Nuclear Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transfection , Wnt Signaling Pathway
9.
Biomed Res Int ; 2018: 6938482, 2018.
Article in English | MEDLINE | ID: mdl-30420966

ABSTRACT

Recent data has shown that cyclin G2 (CCNG2) is an atypical cyclin that inhibits cell cycle progression and is often dysregulated in human cancers. The involvement of cyclin G2 in the occurrence and development of diabetic nephropathy (DN) has not been determined. In the present study, we conducted cyclin G2 knockout studies to determine whether this protein regulates glomerulosclerosis in DN mice. We found that cyclin G2 regulated the expression of renal glomerulosclerosis-related proteins via the canonical Wnt signalling pathway in glomerular mesangial cells. A cyclin G2 deficiency resulted in more severe renal injury in DN mice. These findings provided new insight into the pathogenesis of DN, revealing that cyclin G2 has a protective role in glomerulosclerosis and is a potential new target for the prevention and treatment of DN.


Subject(s)
Cyclin G2/genetics , Cyclin G2/metabolism , Diabetic Nephropathies/metabolism , Animals , Cell Line , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, Knockout , Wnt Signaling Pathway/genetics
10.
Oncol Rep ; 40(4): 2067-2078, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30106441

ABSTRACT

Colorectal cancer (CRC) is among the most common malignancies of the digestive system. Dysregulation of miRNAs and the farnesoid X receptor (FXR) are involved in the progression of CRC. In the present study, the effects of FXR and miR­135A1 in CRC were evaluated. Reverse transcription quantitative­polymerase chain reaction (RT­qPCR) was used to examine the expression of miR­135A1 in patient CRC tissues and adjacent non­tumor tissues, as well as cell lines. The association between miR­135A1 and clinical characteristics of patients with CRC was also investigated. RT­qPCR and western blotting were used to evaluate the expression of miR­135A1 targets. Regulation of cyclin G2 (CCNG2) by miR­135A1was confirmed using luciferase assays. The biological effects of miR­135A1 were assessed in transfected and untransfected CRC cell lines using colony formation assays, cell­cycle analysis by flow cytometry, and CCK­8 assays. miR­135A1 was upregulated in CRC specimens and cell lines. miR­135A1 expression was strongly associated with poor cell differentiation, high expression of carbohydrate antigen (CA)125, CA199, carcinoembryonic antigen and survival rate of patients with CRC. Expression of CCNG2 was downregulated in CRC patients and cell lines, and was further demonstrated to be among the downstream targets of miR­135A1. The present study indicated that inhibition of miR­135A1 expression leads to cell cycle arrest and inhibition of proliferation of CRC cells via increasing CCNG2 expression. In the present study, activation of FXR by GW4064 increased CCNG2 expression via suppression of miR­135A1 expression, and the FXR/miR­135A1/CCNG2 axis was demonstrated to be involved in mediating cell proliferation. In conclusion, activation of FXR by GW4064 suppresses cell proliferation and causes cell cycle arrest in CRC, and the miR­135A1/CCNG2 pathway was suggested to be involved in this step.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Proliferation , Colorectal Neoplasms/pathology , Cyclin G2/metabolism , MicroRNAs/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/surgery , Cyclin G2/genetics , Female , Humans , Male , Middle Aged , Prognosis , Receptors, Cytoplasmic and Nuclear/genetics , Survival Rate , Tumor Cells, Cultured
11.
Cell Physiol Biochem ; 44(5): 1741-1748, 2017.
Article in English | MEDLINE | ID: mdl-29216623

ABSTRACT

BACKGROUND/AIMS: Treatment of breast cancer remains a clinical challenge. This study aims to validate exosomal microRNA-1246 (miR-1246) as a serum biomarker for breast cancer and understand the underlying mechanism in breast cancer progression. METHODS: The expression levels of endogenous and exosomal miRNAs were examined by real time PCR, and the expression level of the target protein was detected by western blot. Scanning electron and confocal microscopy were used to characterize exosomes and to study their uptake and transfer. Luciferase reporter plasmids and its mutant were used to confirm direct targeting. Furthermore, the functional significance of exosomal miR-1246 was estimated by invasion assay and cell viability assay. RESULTS: In this study, we demonstrate that exosomes carrying microRNA can be transferred among different cell lines through direct uptake. miR-1246 is highly expressed in metastatic breast cancer MDA-MB-231 cells compared to non-metastatic breast cancer cells or non-malignant breast cells. Moreover, miR-1246 can suppress the expression level of its target gene, Cyclin-G2 (CCNG2), indicating its functional significance. Finally, treatment with exosomes derived from MDA-MB-231 cells could enhance the viability, migration and chemotherapy resistance of non-malignant HMLE cells. CONCLUSIONS: Together, our results support an important role of exosomes and exosomal miRNAs in regulating breast tumor progression, which highlights their potential for applications in miRNA-based therapeutics.


Subject(s)
Cyclin G2/metabolism , Exosomes/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cluster Analysis , Cyclin G2/antagonists & inhibitors , Cyclin G2/genetics , Drug Resistance, Neoplasm , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , RNA, Small Nuclear/metabolism , Sequence Alignment , Up-Regulation
12.
Sci Rep ; 6: 39091, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27982046

ABSTRACT

Cyclin G1 (CycG1) and Cyclin G2 (CycG2) play similar roles during the DNA damage response (DDR), but their detailed roles remain elusive. To investigate their distinct roles, we generated knockout mice deficient in CycG1 (G1KO) or CycG2 (G2KO), as well as double knockout mice (DKO) deficient in both proteins. All knockouts developed normally and were fertile. Generation of mouse embryonic fibroblasts (MEFs) from these mice revealed that G2KO MEFs, but not G1KO or DKO MEFs, were resistant to DNA damage insults caused by camptothecin and ionizing radiation (IR) and underwent cell cycle arrest. CycG2, but not CycG1, co-localized with γH2AX foci in the nucleus after γ-IR, and γH2AX-mediated DNA repair and dephosphorylation of CHK2 were delayed in G2KO MEFs. H2AX associated with CycG1, CycG2, and protein phosphatase 2A (PP2A), suggesting that γH2AX affects the function of PP2A via direct interaction with its B'γ subunit. Furthermore, expression of CycG2, but not CycG1, was abnormal in various cancer cell lines. Kaplan-Meier curves based on TCGA data disclosed that head and neck cancer patients with reduced CycG2 expression have poorer clinical prognoses. Taken together, our data suggest that reduced CycG2 expression could be useful as a novel prognostic marker of cancer.


Subject(s)
Cyclin G1/genetics , Cyclin G2/genetics , Fibroblasts/cytology , Head and Neck Neoplasms/genetics , Animals , Camptothecin/adverse effects , Cell Line, Tumor , Cells, Cultured , Checkpoint Kinase 2/metabolism , Cyclin G1/metabolism , Cyclin G2/metabolism , DNA Damage , DNA Repair , Down-Regulation , Fibroblasts/drug effects , Fibroblasts/radiation effects , Head and Neck Neoplasms/metabolism , Mice , Mice, Knockout , Phenotype , Phosphorylation , Radiation, Ionizing
13.
Oncol Rep ; 36(2): 1111-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27374211

ABSTRACT

Aberrant expression and function of microRNAs (miRNAs) play a critical role in the development and progression of various human cancers including gastric cancer. However, the clinical significance and underlying mechanisms of miR-340 remain largely unknown in gastric cancer. In the present study, we demonstrated that the expression of miR-340 was aberrantly elevated in both gastric cancer tissues and cells. Moreover clinical association analyses disclosed that the elevated level of miR-340 was significantly associated with unfavorable clinicopathological characteristics of the gastric cancer patients, such as poor differentiation, large tumor size and advanced tumor-node-metastasis (TNM) stage. Gastric cancer patients with high expression of miR-340 had prominently shorter overall survival and disease-free survival. Functionally, forced expression of miR-340 promoted cell viability, proliferation, colony formation and cell cycle progression in the SGC-7901 cells, while miR-340 silencing reduced cell viability, proliferation, colony formation and cell cycle progression in MGC-803 cells. Furthermore, in vivo experiments indicated that miR-340 knockdown suppressed the tumor growth of MGC-803 cells. Notably, alteration of miR-340 expression affected the luciferase activity of wild-type 3'-UTR of cyclin G2 (CCNG2) and regulated CCNG2 abundance in gastric cancer cells, indicating that CCNG2 is a direct target of miR-340. Moreover, CCNG2 knockdown eradicated the effects of miR-340 silencing on gastric cancer cells. In conclusion, our data suggest that miR-340 may potentially serve as a novel prognostic biomarker and therapeutic target for gastric cancer.


Subject(s)
Cell Proliferation/genetics , Cyclin G2/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , Tumor Burden/genetics
14.
Oncogene ; 35(36): 4816-27, 2016 09 08.
Article in English | MEDLINE | ID: mdl-26876206

ABSTRACT

Epithelial ovarian cancer (EOC) has the highest mortality rate among gynecological malignancies owing to poor screening methods, non-specific symptoms and limited knowledge of the cellular targets that contribute to the disease. Cyclin G2 is an unconventional cyclin that acts to oppose cell cycle progression. Dysregulation of the cyclin G2 gene (CCNG2) in a variety of human cancers has been reported; however, the role of cyclin G2 in tumorigenesis remains unclear. In this study, we investigated the function of cyclin G2 in EOC. In vitro and in vivo studies using several EOC-derived tumor cell lines revealed that cyclin G2 inhibited cell proliferation, migration, invasion and spheroid formation, as well as tumor formation and invasion. By interrogating cDNA microarray data sets, we found that CCGN2 mRNA is reduced in several large cohorts of human ovarian carcinoma when compared with normal ovarian surface epithelium or borderline tumors of the ovary. Mechanistically, cyclin G2 was found to suppress epithelial-to-mesenchymal transition (EMT), as demonstrated by the differential regulation of various EMT genes, such as Snail, Slug, vimentin and E-cadherin. Moreover, cyclin G2 potently suppressed the Wnt/ß-catenin signaling pathway by downregulating key Wnt components, namely LRP6, DVL2 and ß-catenin, which could be linked to inhibition of EMT. Taken together, our novel findings demonstrate that cyclin G2 has potent tumor-suppressive effects in EOCs by inhibiting EMT through attenuating Wnt/ß-catenin signaling.


Subject(s)
Carcinogenesis/genetics , Cyclin G2/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Antigens, CD , Cadherins/genetics , Carcinoma, Ovarian Epithelial , Cell Movement/genetics , Cell Proliferation/genetics , Dishevelled Proteins/antagonists & inhibitors , Dishevelled Proteins/genetics , Female , Humans , Low Density Lipoprotein Receptor-Related Protein-6/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Neoplasm Invasiveness/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Wnt Signaling Pathway/genetics , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
15.
Int J Oncol ; 47(2): 473-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26046133

ABSTRACT

Although all-trans retinoic acid (ATRA) is a standard and effective drug used for differentiation therapy in acute promyelocytic leukemia, ATRA-resistant leukemia cells ultimately emerge during this treatment. Therefore, the development of new drugs or effective combination therapy is urgently needed. We demonstrate that the combined treatment of vitamin K2 and cotylenin A synergistically induced monocytic differentiation in HL-60 cells. This combined treatment also synergistically induced NBT-reducing activity and non-specific esterase-positive cells as well as morphological changes to monocyte/macrophage-like cells. Vitamin K2 and cotylenin A cooperatively inhibited the proliferation of HL-60 cells in short-term and long-term cultures. This treatment also induced growth arrest at the G1 phase. Although 5 µg/ml cotylenin A or 5 µM vitamin K2 alone reduced c-MYC gene expression in HL-60 cells to approximately 45% or 80% that of control cells, respectively, the combined treatment almost completely suppressed c-MYC gene expression. We also demonstrated that the combined treatment of vitamin K2 and cotylenin A synergistically induced the expression of cyclin G2, which had a positive effect on the promotion and maintenance of cell cycle arrest. These results suggest that the combination of vitamin K2 and cotylenin A has therapeutic value in the treatment of acute myeloid leukemia.


Subject(s)
Cyclin G2/genetics , Diterpenes/pharmacology , Leukemia/genetics , Monocytes/drug effects , Proto-Oncogene Proteins c-myc/genetics , Vitamin K 2/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Leukemia/drug therapy , Monocytes/cytology
16.
Leukemia ; 29(10): 1993-2002, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25925206

ABSTRACT

The BMI1 polycomb protein regulates self-renewal, proliferation and survival of cancer-initiating cells essentially through epigenetic repression of the CDKN2A tumor suppressor locus. We demonstrate here for the first time that BMI1 also prevents autophagy in chronic myeloid leukemia (CML) cell lines, to support their proliferation and clonogenic activity. Using chromatin immunoprecipitation, we identified CCNG2/cyclin G2 (CCNG2) as a direct BMI1 target. BMI1 downregulation in CD34+ CML cells by PTC-209 pharmacological treatment or shBMI1 transduction triggered CCNG2 expression and decreased clonogenic activity. Also, ectopic expression of CCNG2 in CD34+ CML cells strongly decreased their clonogenicity. CCNG2 was shown to act by disrupting the phosphatase 2A complex, which activates a PKCζ-AMPK-JNK-ERK pathway that engages autophagy. We observed that BMI1 and CCNG2 levels evolved inversely during the progression of CML towards an acute deadly phase, and therefore hypothesized that BMI1 could support acute transformation of CML through the silencing of a CCNG2-mediated tumor-suppressive autophagy response.


Subject(s)
Autophagy , Cell Proliferation , Cyclin G2/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Polycomb Repressive Complex 1/metabolism , Apoptosis , Blotting, Western , Chromatin Immunoprecipitation , Cyclin G2/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured
17.
Mol Cancer ; 14: 36, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25888956

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are short, non-coding RNA molecules that play critical roles in human malignancy. However, the regulatory characteristics of miRNAs in triple-negative breast cancer, a phenotype of breast cancer that does not express the genes for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2, are still poorly understood. METHODS: In this study, miRNA expression profiles of 24 triple-negative breast cancers and 14 adjacent normal tissues were analyzed using deep sequencing technology. Expression levels of miRNA reads were normalized with the quantile-quantile scaling method. Deregulated miRNAs in triple-negative breast cancer were identified from the sequencing data using the Student's t-test. Quantitative reverse transcription PCR validations were carried out to examine miRNA expression levels. Potential target candidates of a miRNA were predicted using published target prediction algorithms. Luciferase reporter assay experiments were performed to verify a putative miRNA-target relationship. Validated molecular targets of the deregulated miRNAs were retrieved from curated databases and their associations with cancer progression were discussed. RESULTS: A novel 25-miRNA expression signature was found to effectively distinguish triple-negative breast cancers from surrounding normal tissues in a hierarchical clustering analysis. We documented the evidence of seven polycistronic miRNA clusters preferentially harboring deregulated miRNAs in triple-negative breast cancer. Two of these miRNA clusters (miR-143-145 at 5q32 and miR-497-195 at 17p13.1) were markedly down-regulated in triple-negative breast cancer, while the other five miRNA clusters (miR-17-92 at 13q31.3, miR-183-182 at 7q32.2, miR-200-429 at 1p36.33, miR-301b-130b at 22q11.21, and miR-532-502 at Xp11.23) were up-regulated in triple-negative breast cancer. Moreover, miR-130b-5p from the miR-301b-130b cluster was shown to directly repress the cyclin G2 (CCNG2) gene, a crucial cell cycle regulator, in triple-negative breast cancer cells. Luciferase reporter assays showed that miR-130b-5p-mediated repression of CCNG2 was dependent on the sequence of the 3'-untranslated region. The findings described in this study implicate a miR-130b-5p-CCNG2 axis that may be involved in the malignant progression of triple-negative breast cancer. CONCLUSIONS: Our work delivers a clear picture of the global miRNA regulatory characteristics in triple-negative breast cancer and extends the current knowledge of microRNA regulatory network.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Transcriptome/genetics , Triple Negative Breast Neoplasms/genetics , 3' Untranslated Regions/genetics , Adult , Aged , Aged, 80 and over , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cyclin G2/genetics , Down-Regulation/genetics , Female , HEK293 Cells , High-Throughput Nucleotide Sequencing/methods , Humans , Middle Aged , Up-Regulation/genetics
18.
Int J Oncol ; 46(1): 161-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25309979

ABSTRACT

microRNA93 (miR-93) is expressed in the miR­106b-25 cluster, located in intron 13 of the MCM7 gene. Our previous study found that miR-93 was significantly upregulated in laryngeal squamous cell carcinoma (LSCC), and cyclin G2 (CCNG2) was a potential target of miR-93 in LSCC. However, the possible functions and molecular mechanisms of miR-93 in LSCC remain unknown. In the present study, we show that the level of CCNG2 protein expression was significantly lower in LSCC cancer tissue than normal tissues. The level of CCNG2 was correlated with clinical stages, lymph node metastasis and histological grade. We further show that the expression level of miR-93 was inversely correlated with CCNG2 expression in clinical specimens. Furthermore, gain-of-function assays revealed that miR-93 promoted cell proliferation, decreased apoptosis rates, induced cell cycle arrest and promoted cell migration and invasion, whereas silencing of miR-93 attenuated these carcinogenic processes. In addition, overexpression of miR-93 in Hep-2 cells could reduce the mRNA and protein levels of CCNG2, whereas silencing of miR-93 in Hep-2 cells significantly increased CCNG2 expression. A luciferase assay verified that miR-93 could bind to the 3' untranslated region of CCNG2. Importantly, ectopic expression of CCNG2 in miR-93 cells rescued the effect of miR-93 on LSCC proliferation. Knockdown of CCNG2 promoted cell proliferation resembling that of miR-93 overexpression. These findings demonstrated that miR-93 promotes tumor growth by directly suppressing CCNG2. Taken together, these results suggested that this newly identified miR-93-CCNG2 axis may be involved in LSCC proliferation and progression. Our findings provide novel potential targets for LSCC therapy and prognosis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cyclin G2/genetics , Laryngeal Neoplasms/genetics , MicroRNAs/physiology , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Squamous Cell/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Laryngeal Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Oncogenes , Tumor Cells, Cultured
19.
Biol Reprod ; 91(3): 76, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25122062

ABSTRACT

MicroRNAs are expressed abundantly in the placenta throughout pregnancy. We have previously reported that microRNA (miR)-378a-5p promoted trophoblast migration and invasion. To further understand the role of miR-378a-5p during placental development, we investigated whether it may regulate the differentiation of syncytiotrophoblast (STB). Using a choriocarcinoma cell line, BeWo, we found that miR-378a-5p was down-regulated during forskolin-induced STB differentiation. Transfection of a miR-378a-5p mimic into BeWo cells decreased the formation of multinucleated STB, increased E-cadherin, and decreased the expression level of STB marker genes. On the other hand, transfection of anti-miR-378a-5p resulted in an increase in formation of multinucleated STB and expression of STB marker genes, as well as the loss of E-cadherin. Bioinformatic analysis revealed that miR-378a-5p has four potential binding sites at the 3' untranslated region (UTR) of cyclin G2 (CCNG2). Using luciferase reporter assays, we showed that miR-378a-5p decreased the luciferase activity of reporter constructs that contain CCNG2 3' UTR. In addition, miR-378a-5p decreased, whereas anti-miR-378a-5p increased, CCNG2 mRNA levels. Overexpression of CCNG2 increased the expression of syncytin-1 and fusion index and reversed the inhibitory effects of miR-378a-5p. In contrast, silencing of CCNG2 using siRNA increased E-cadherin and decreased syncytin-1 levels. These findings provide initial evidence that CCNG2 promotes STB differentiation and suggest that miR-378a-5p exerts an inhibitory role in STB differentiation, in part, by down-regulating CCNG2 expression, in the BeWo cell model.


Subject(s)
Cell Differentiation , Cyclin G2/antagonists & inhibitors , Down-Regulation , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Placentation , Trophoblasts/metabolism , 3' Untranslated Regions , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Choriocarcinoma/genetics , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Cyclin G2/genetics , Cyclin G2/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/chemistry , MicroRNAs/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pregnancy , RNA/antagonists & inhibitors , RNA/genetics , RNA/metabolism , Response Elements , Transfection , Trophoblasts/pathology
20.
Surg Obes Relat Dis ; 10(4): 691-6, 2014.
Article in English | MEDLINE | ID: mdl-24708911

ABSTRACT

BACKGROUND: The involvement of cyclin G2 (CCNG2) and cyclin-dependent kinase-4 (CDK4), cell cycle regulatory proteins, in adipose tissue metabolism and insulin resistance is still unknown. The objective of this study was to analyze CCNG2 and CDK4 levels in visceral (VAT) and subcutaneous adipose tissue (SAT) from nonobese and morbidly obese patients and their relationship with insulin resistance. METHODS: We studied the mRNA and protein levels of CCNG2 and CDK4 in VAT and SAT from 12 nonobese and 23 morbidly obese patients (11 with low [MO-L-IR] and 12 with high insulin resistance [MO-H-IR]). RESULTS: The nonobese patients had a significantly greater CCNG2 expression in VAT (P = .004) and SAT (P<.001) than the MO-L-IR and MO-H-IR patients. The MO-H-IR patients had a significantly lower CDK4 expression in VAT than the MO-L-IR (P = .026), but similar to the nonobese patients. CDK4 and CCNG2 expression correlated significantly in VAT (r = 0.511, P<.001) and SAT (r = .535, P = .001). In different multiple regression analysis models, CCNG2 and CDK4 expression in VAT was mainly predicted by glucose (P = .047 and P = .008, respectively), and CCNG2 expression in SAT was mainly predicted by body mass index (P = .041). No significant associations were found with CDK4 expression in SAT. Moreover, VAT CCNG2 expression was the main determinant of the improvement in the homeostasis model assessment of insulin resistance index at 3 months after bariatric surgery (B = -271.7, P = .026). CONCLUSION: Our data show for the first time that the human CCNG2 and CDK4 expression of VAT are inversely associated with glucose and insulin resistance.


Subject(s)
Cyclin G2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Insulin Resistance/physiology , Intra-Abdominal Fat/metabolism , Obesity, Morbid/metabolism , Subcutaneous Fat/metabolism , Adult , Bariatric Surgery , Body Mass Index , Case-Control Studies , Cyclin G2/genetics , Cyclin-Dependent Kinase 4/genetics , Female , Humans , Male , Middle Aged , Obesity, Morbid/surgery , Prospective Studies , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...