Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
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
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.
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
4.
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
5.
Nature ; 487(7407): 380-4, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22801492

ABSTRACT

The molecular determinants of malignant cell behaviours in breast cancer remain only partially understood. Here we show that SHARP1 (also known as BHLHE41 or DEC2) is a crucial regulator of the invasive and metastatic phenotype in triple-negative breast cancer (TNBC), one of the most aggressive types of breast cancer. SHARP1 is regulated by the p63 metastasis suppressor and inhibits TNBC aggressiveness through inhibition of hypoxia-inducible factor 1α (HIF-1α) and HIF-2α (HIFs). SHARP1 opposes HIF-dependent TNBC cell migration in vitro, and invasive or metastatic behaviours in vivo. SHARP1 is required, and sufficient, to limit expression of HIF-target genes. In primary TNBC, endogenous SHARP1 levels are inversely correlated with those of HIF targets. Mechanistically, SHARP1 binds to HIFs and promotes HIF proteasomal degradation by serving as the HIF-presenting factor to the proteasome. This process is independent of pVHL (von Hippel-Lindau tumour suppressor), hypoxia and the ubiquitination machinery. SHARP1 therefore determines the intrinsic instability of HIF proteins to act in parallel to, and cooperate with, oxygen levels. This work sheds light on the mechanisms and pathways by which TNBC acquires invasiveness and metastatic propensity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cyclin G2/genetics , Female , Humans , Kaplan-Meier Estimate , Multivariate Analysis
6.
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
7.
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
8.
Biochem J ; 457(1): 69-77, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24059861

ABSTRACT

The mechanisms whereby insulin analogues may cause enhanced mitogenicity through activation of either the IR (insulin receptor) or the IGF-IR (insulin-like growth factor 1 receptor) are incompletely understood. We demonstrate that in L6 myoblasts expressing only IGF-IRs as well as in the same cells overexpressing the IR, IGF-I (insulin-like growth factor 1), insulin and X10 (AspB10 insulin) down-regulate the mRNA expression level of the cell cycle inhibitor cyclin G2, as measured by qRT-PCR (quantitative reverse transcription-PCR), and induce cell growth measured by [6-(3)H]thymidine incorporation into DNA. Western blotting showed a marked down-regulation of cyclin G2 at the protein level in both cell lines. Overexpression of cyclin G2 in the two cell lines diminished the mitogenic effect of all three ligands. The use of specific inhibitors indicated that both the MAPK (mitogen-activated protein kinase) and the PI3K (phosphoinositide 3-kinase) pathways mediate the down-regulation of Ccng2. The down-regulation of CCNG2 by the three ligands was also observed in other cell lines: MCF-7, HMEC, Saos-2, R(-)/IR and INS-1. These results indicate that regulation of cyclin G2 is a key mechanism whereby insulin, insulin analogues and IGF-I stimulate cell proliferation.


Subject(s)
Cyclin G2/genetics , Insulin-Like Growth Factor I/pharmacology , Insulin/analogs & derivatives , Mitosis , Cell Proliferation/drug effects , Cells, Cultured , DNA/biosynthesis , Down-Regulation/drug effects , Humans , Insulin/pharmacology , MCF-7 Cells , Mitosis/drug effects , Mitosis/physiology , Peptides/pharmacology , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
9.
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
10.
Tumour Biol ; 35(4): 3339-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24307622

ABSTRACT

This study aimed to analyze the expression, clinical significance of cyclin G2 (CCNG2) in colorectal carcinoma, and the biological effect in its cell line by CCNG2 overexpression. Immunohistochemistry and Western blot were used to analyze CCNG2 protein expression in colorectal cancer and to study the influence of the upregulated expression of CCNG2 that might be found on SW480 cell biological effect. We found that the level of CCNG2 protein expression was significantly lower in colorectal cancer tissue than normal tissues (P < 0.05). The level of CCNG2 was correlated with T stages, lymph node metastasis, clinic stage, and histological grade (P < 0.05). Loss of CCNG2 expression correlated significantly with poor overall survival time by Kaplan-Meier analysis (P < 0.05). The result of biological function has shown that SW480 cell-transfected CCNG2 had a lower survival fraction, higher percentage of the G0/G1 phases, and lower CDK2 protein expression compared with SW480 cell-untransfected CCNG2 (P < 0.05).


Subject(s)
Colorectal Neoplasms/pathology , Cyclin G2/physiology , Adult , Aged , Cell Cycle , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Cyclin G2/analysis , Cyclin G2/genetics , Cyclin-Dependent Kinase 2/physiology , Down-Regulation , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis
11.
Tumour Biol ; 35(4): 3355-62, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24297335

ABSTRACT

This study aimed to analyze the expression, clinical significance of cyclin G2 (CCNG2) in esophageal carcinoma, and the biological effect in its cell line by CCNG2 overexpression. Immunohistochemistry and western blot were used to analyze CCNG2 protein expression in 73 cases of esophageal cancer and normal tissues to study the relationship between CCNG2 expression and clinical factors. CCNG2 lentiviral vector and empty vector were respectively transfected into esophageal cancer Eca-109 cell line. Reverse transcription-polymerase chain reaction and western blot were used to detect the mRNA level and protein of CCNG2. MTT assay and cell cycle were also conducted as to the influence of the upregulated expression of CCNG2 that might be found on Eca-109 cell's biological effect. Immunohistochemistry: The level of CCNG2 protein expression was found to be significantly lower in esophageal cancer tissue than normal tissues (P < 0.05). Western blot: The relative amount of CCNG2 protein in esophageal cancer tissue was respectively found to be significantly lower than in normal tissues (P < 0.05). The level of CCNG2 protein expression was not correlated with gender, age, and tumor size (P > 0.05), but it was correlated with lymph node metastasis, clinic stage, and histological grades (P < 0.05). Loss of CCNG2 expression correlated significantly with poor overall survival time by Kaplan-Meier analysis. The result of the biological function showed that Eca-109 cell-transfected CCNG2 had a lower survival fraction, more percentage of the G0/G1 phases (P < 0.05), and lower cyclin-dependent kinase 2 (CDK2) protein expression. CCNG2 expression decreased in esophageal cancer and correlated significantly with lymph node metastasis, clinic stage, histological grade, and poor overall survival, suggesting that CCNG2 may play important roles as a negative regulator to esophageal cancer cell by promoting degradation of CDK2.


Subject(s)
Cyclin G2/physiology , Esophageal Neoplasms/pathology , Adult , Aged , Cyclin G2/analysis , Cyclin G2/genetics , Cyclin-Dependent Kinase 2/analysis , Esophageal Neoplasms/chemistry , Esophageal Neoplasms/mortality , Esophagus/chemistry , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis
12.
Tumour Biol ; 35(4): 3177-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24272084

ABSTRACT

This study aims to analyze the expression and clinical significance of cyclin G2 (CCNG2) in kidney carcinoma, and the biological effect in its cell line by CCNG2 overexpression. Immunohistochemistry and western blot were used to analyze CCNG2 protein expression in 63 cases of kidney cancer and normal tissues to study the relationship between CCNG2 expression and clinical factors. CCNG2 lentiviral vector and empty vector were respectively transfected into kidney ACHN cell line. During immunohistochemistry, the level of CCNG2 protein expression was found to be significantly lower in kidney cancer tissue than normal tissues (P < 0.05). After Western blot, the relative amount of CCNG2 protein in kidney cancer tissue was respectively found to be significantly lower than in normal tissues (P < 0.05). The level of CCNG2 protein expression was not correlated with gender, age, tumor size, and pathological types (P > 0.05), but it was correlated with lymph node metastasis, clinic stage, and histological grade (P < 0.05). Loss of CCNG2 expression correlated significantly with poor overall survival time by Kaplan-Meier analysis (P < 0.05). The result of biological function show that ACHN cell-transfected CCNG2 had a lower survival fraction, higher percentage of the G0/G1 phases, and lower CDK2 protein expression compared with ACHN cell-untransfected CCNG2 (P < 0.05). CCNG2 expression decreased in kidney cancer and correlated significantly with lymph node metastasis, clinical stage, histological grade, and poor overall survival, suggesting that CCNG2 may play important roles as a negative regulator to kidney cancer ACHN cell by promoting degradation of CDK2.


Subject(s)
Cyclin G2/physiology , Kidney Neoplasms/pathology , Adult , Aged , Cell Line, Tumor , Cyclin G2/analysis , Cyclin G2/genetics , Cyclin-Dependent Kinase 2/metabolism , Female , Humans , Immunohistochemistry , Kidney/chemistry , Kidney Neoplasms/chemistry , Kidney Neoplasms/mortality , Lymphatic Metastasis , Male , Middle Aged , Prognosis
13.
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
14.
J Biol Chem ; 287(27): 22838-53, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22589537

ABSTRACT

To maintain genomic integrity DNA damage response (DDR), signaling pathways have evolved that restrict cellular replication and allow time for DNA repair. CCNG2 encodes an unconventional cyclin homolog, cyclin G2 (CycG2), linked to growth inhibition. Its expression is repressed by mitogens but up-regulated during cell cycle arrest responses to anti-proliferative signals. Here we investigate the potential link between elevated CycG2 expression and DDR signaling pathways. Expanding our previous finding that CycG2 overexpression induces a p53-dependent G(1)/S phase cell cycle arrest in HCT116 cells, we now demonstrate that this arrest response also requires the DDR checkpoint protein kinase Chk2. In accord with this finding we establish that ectopic CycG2 expression increases phosphorylation of Chk2 on threonine 68. We show that DNA double strand break-inducing chemotherapeutics stimulate CycG2 expression and correlate its up-regulation with checkpoint-induced cell cycle arrest and phospho-modification of proteins in the ataxia telangiectasia mutated (ATM) and ATM and Rad3-related (ATR) signaling pathways. Using pharmacological inhibitors and ATM-deficient cell lines, we delineate the DDR kinase pathway promoting CycG2 up-regulation in response to doxorubicin. Importantly, RNAi-mediated blunting of CycG2 attenuates doxorubicin-induced cell cycle checkpoint responses in multiple cell lines. Employing stable clones, we test the effect that CycG2 depletion has on DDR proteins and signals that enforce cell cycle checkpoint arrest. Our results suggest that CycG2 contributes to DNA damage-induced G(2)/M checkpoint by enforcing checkpoint inhibition of CycB1-Cdc2 complexes.


Subject(s)
Cell Division/physiology , Cyclin G2/genetics , DNA Damage/physiology , Doxorubicin/pharmacology , G2 Phase/physiology , Signal Transduction/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Ataxia Telangiectasia Mutated Proteins , CDC2 Protein Kinase , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Checkpoint Kinase 2 , Cyclin B/metabolism , Cyclin B1/metabolism , Cyclin G2/metabolism , Cyclin-Dependent Kinases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , G2 Phase/drug effects , Genes, cdc/drug effects , Genes, cdc/physiology , HCT116 Cells , Humans , Mice , NIH 3T3 Cells , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/physiopathology , Oncogene Protein p21(ras)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL