Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chem Biol Interact ; 387: 110809, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38006958

RESUMEN

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.


Asunto(s)
Ciclina G2 , MicroARNs , Humanos , Ciclina G2/genética , Ciclina G2/metabolismo , Fase S , Hidroquinonas/toxicidad , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Transformación Celular Neoplásica
2.
J Exp Clin Cancer Res ; 41(1): 358, 2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36566226

RESUMEN

BACKGROUND: IFN-γ is a key mediator of tumor immunity that can induce macrophage polarization to suppress tumor growth. Cyclin G2 functions as a tumor suppressor in various cancer cells; however, its role in macrophages remains unclear. This study aimed to investigate the role and underlying mechanisms of cyclin G2 in macrophages in vitro and in vivo. METHODS: Mouse tumor models were used to determine the effect of cyclin G2 in macrophages on tumor growth in vivo following IFN-γ treatment. Immunohistochemistry staining, immunofluorescence staining and flow cytometry were used to evaluate the number of cytotoxic T lymphocytes (CTLs) and blood vessels in the mouse tumors. Moreover, the biological roles of cyclin G2 in macrophages with regard to CTL chemotaxis, cytotoxic function, and vascular endothelial cell tube formation were assessed using in vitro functional experiments. Immunoprecipitation (IP), real-time PCR, and enzyme-linked immunosorbent assays (ELISAs) were conducted to investigate the underlying mechanisms by which cyclin G2 regulates CTLs and vascular endothelial cells. RESULTS: We found that cyclin G2 expression was upregulated in macrophages after IFN-γ treatment. Upregulated cyclin G2 inhibited lung and colon cancer growth by increasing the secretion of its downstream effector CXCL9, which promoted CTL chemotaxis and suppressed vascular endothelial cell tube formation. Moreover, cyclin G2 increased CXCL9 mRNA levels by promoting STAT1 nuclear translocation. In addition, cyclin G2 promoted the activation of the STAT1 signaling pathway, which was dependent on PP2Ac. CONCLUSIONS: Cyclin G2 is upregulated by IFN-γ in macrophages, promotes the secretion of CXCL9 to increase CTL chemotaxis and inhibit angiogenesis to suppress tumor growth. Our findings suggest that targeting cyclin G2 could benefit future immunotherapy.


Asunto(s)
Ciclina G2 , Interferón gamma , Macrófagos , Neoplasias , Neovascularización Patológica , Linfocitos T Citotóxicos , Animales , Ratones , Línea Celular Tumoral , Ciclina G2/metabolismo , Células Endoteliales/metabolismo , Inmunoterapia , Interferón gamma/metabolismo , Macrófagos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neovascularización Patológica/metabolismo
3.
BMC Cancer ; 21(1): 1074, 2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34598688

RESUMEN

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.


Asunto(s)
Ciclina G2/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Neoplasias de Cabeza y Cuello/metabolismo , Puntos de Control de la Fase M del Ciclo Celular , MicroARNs/metabolismo , Recurrencia Local de Neoplasia/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Regiones no Traducidas 3'/genética , Apoptosis/genética , Área Bajo la Curva , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Ciclina G2/genética , Regulación hacia Abajo , Femenino , Silenciador del Gen , Neoplasias de Cabeza y Cuello/genética , Humanos , Luciferasas/metabolismo , Masculino , MicroARNs/genética , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , ARN Mensajero/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Transfección , Regulación hacia Arriba
4.
J Exp Clin Cancer Res ; 40(1): 273, 2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34452627

RESUMEN

BACKGROUND: Expression of aberrant cyclin G2 is a key factor contributing to cancer biological processes, including glioma. However, the potential underlying mechanisms of cyclin G2 in the glioma tumor immune microenvironment remain unclear. METHODS: Co-immunoprecipitation (co-IP), in situ proximity ligation assay (PLA), and in vitro kinase assay were conducted to reveal the underlying mechanism by which cyclin G2 regulates Y10 phosphorylation of LDHA. Further, the biological roles of cyclin G2 in cell proliferation, migration, invasion capacity, apoptosis, glycolysis, and immunomodulation were assessed through in vitro and in vivo functional experiments. Expressions of cyclin G2 and Foxp3 in glioma specimens was determined by immunohistochemistry. RESULTS: In this study, we found that cyclin G2 impeded the interaction between LDHA and FGFR1, thereby decreasing Y10 phosphorylation of LDHA through FGFR1 catalysis. Cyclin G2 inhibited proliferation, migration, invasion capacity, and glycolysis and promoted apoptosis glioma cells via suppressing Y10 phosphorylation of LDHA. Moreover, we further verified that cyclin G2 reversed the immunosuppressive to antitumor immune microenvironment through inhibiting lactate production by glioma cells. Besides, cyclin G2 potentiated PD-1 blockade and exerted strong antitumor immunity in the glioma-bearing mice model. CONCLUSIONS: Cyclin G2 acts as a potent tumor suppressor in glioma and enhances responses to immunotherapy. Our findings may be helpful in selecting glioma patients for immunotherapy trials in the future.


Asunto(s)
Neoplasias Encefálicas/patología , Ciclina G2/metabolismo , Glioma/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Microambiente Tumoral/inmunología , Animales , Apoptosis , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Glioma/metabolismo , Glucólisis , Humanos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Invasividad Neoplásica , Fosforilación
5.
FASEB J ; 35(2): e21169, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33205477

RESUMEN

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.


Asunto(s)
Movimiento Celular/genética , Ciclina G1/metabolismo , Ciclina G2/metabolismo , Proteínas Dishevelled/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Preeclampsia/metabolismo , Trofoblastos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba/genética , Adulto , Animales , Línea Celular , Ciclina G1/genética , Ciclina G2/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Preeclampsia/genética , Preeclampsia/patología , Embarazo , Transfección , Ubiquitina-Proteína Ligasas/genética
6.
Med Sci Monit ; 26: e926414, 2020 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-32941407

RESUMEN

BACKGROUND Remodeling of maternal spiral arteries after embryo implantation relies on well-regulated trophoblast functions. Although cyclin G2 (CCNG2) is thought to be involved in placental development and function, its role in trophoblasts and the mechanisms underlying placental development and function remain unclear. The present study investigated the potential role of CCNG2 in trophoblast cell proliferation and their interactions with endothelial cells. MATERIAL AND METHODS CCNG2 levels were modified by stable infection of HTR8/SVneo cells with lentiviruses overexpressing and silencing CCNG2. Cell proliferation was measured using CCK-8 assays. Network formation assays were performed using trophoblasts alone and co-cultured trophoblasts and endothelial cells to measure angiogenesis of trophoblasts and trophoblast-endothelial interactions. Levels of angiogenic factors (VEGF and sFlt-1) in the supernatant were measured by ELISA, and the expression of cell cycle regulatory (cyclin D1) and invasive (MMP2, MMP3, MMP9) markers implicated in artery remodeling were measured by western blotting. RESULTS Ectopic expression of CCNG2 blocked the proliferation of HTR8/SVneo cells, as well as their abilities to form networks and integrate into human umbilical vein endothelial cells, whereas CCNG2 inhibition had the opposite effects. CCNG2 upregulation significantly reduced the expression of VEGF, cyclin D1, MMP2, MMP3, and MMP9, but enhanced the expression of sFlt-1. In contrast, CCNG2 downregulation had the opposite effects. CONCLUSIONS CCNG2 plays a critical role in trophoblast proliferation and trophoblast-endothelial cell interactions by significant affecting cell cycle, angiogenic, and invasive markers. CCNG2 may thus be a novel marker for the treatment of placental disorders.


Asunto(s)
Comunicación Celular , Proliferación Celular , Ciclina G2/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Trofoblastos/metabolismo , Línea Celular , Humanos
7.
IUBMB Life ; 72(7): 1491-1503, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32196930

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinogénesis/patología , Carcinoma de Células Escamosas/patología , Ciclina G2/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas/patología , MicroARNs/genética , Anciano , Apoptosis , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Movimiento Celular , Proliferación Celular , Ciclina G2/genética , Humanos , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Invasividad Neoplásica , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
8.
J Cell Mol Med ; 24(5): 2749-2760, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31978940

RESUMEN

Cyclin G2 (CCNG2) is an atypical cyclin that inhibits cell cycle progression and is often dysregulated in human cancers. Cyclin G2 in the occurrence and development of diabetic nephropathy (DN), one of the most severe diabetic complications, has not been fully identified. In this study, we investigated the function and regulatory mechanism of cyclin G2 in DN. In vivo studies revealed that a deficiency of cyclin G2 significantly increased albuminuria and promoted tubulointerstitial fibrosis in established DN. Cyclin G2 regulated the expression of fibrosis-related proteins via the canonical Wnt signalling pathway in renal tubular epithelial cells. Moreover, the binding of cyclin G2 to Dapper1 (Dpr1/DACT1), a protein involved in Wnt signalling, decreased the phosphorylation of Dpr1 at Ser762 by casein kinase 1 (CK1) and suppressed the Wnt signalling pathway. These findings reveal that cyclin G2 can protect against renal injury and fibrosis associated with DN and, thus, is a new target for the prevention and treatment of diabetic complications.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ciclina G2/metabolismo , Túbulos Renales/patología , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Vía de Señalización Wnt , Albuminuria/complicaciones , Albuminuria/genética , Animales , Quinasa de la Caseína I/metabolismo , Ciclina G2/deficiencia , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Fibrosis , Glucosa/toxicidad , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Índice de Severidad de la Enfermedad
9.
Cancer Lett ; 452: 158-167, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-30922918

RESUMEN

PTOV1 is a transcription and translation regulator and a promoter of cancer progression. Its overexpression in prostate cancer induces transcription of drug resistance and self-renewal genes, and docetaxel resistance. Here we studied PTOV1 ability to directly activate the transcription of ALDH1A1 and CCNG2 by binding to specific promoter sequences. Chromatin immunoprecipitation and electrophoretic mobility shift assays identified a DNA-binding motif inside the PTOV-A domain with similarities to known AT-hooks that specifically interacts with ALDH1A1 and CCNG2 promoters. Mutation of this AT-hook-like sequence significantly decreased the expression of ALDH1A1 and CCNG2 promoted by PTOV1. Immunohistochemistry revealed the association of PTOV1 with mitotic chromosomes in high grade prostate, colon, bladder, and breast carcinomas. Overexpression of PTOV1, ALDH1A1, and CCNG2 significantly correlated with poor prognosis in prostate carcinomas and with shorter relapse-free survival in colon carcinoma. The previously described interaction with translation complexes and its direct binding to ALDH1A1 and CCNG2 promoters found here reveal the PTOV1 capacity to modulate the expression of critical genes at multiple levels in aggressive cancers. Remarkably, the AT-hook motifs in PTOV1 open possibilities for selective targeting its nuclear and/or cytoplasmic activities.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/metabolismo , Biomarcadores de Tumor/genética , Ciclina G2/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/patología , Retinal-Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1/biosíntesis , Línea Celular Tumoral , Ciclina G2/biosíntesis , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Humanos , Masculino , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/genética , Retinal-Deshidrogenasa/biosíntesis
10.
Int J Biol Sci ; 15(3): 544-555, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30745841

RESUMEN

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.


Asunto(s)
Proliferación Celular/fisiología , L-Lactato Deshidrogenasa/metabolismo , Cicatrización de Heridas/fisiología , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/genética , Ciclina G2/genética , Ciclina G2/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , L-Lactato Deshidrogenasa/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Cicatrización de Heridas/genética
11.
J Exp Clin Cancer Res ; 37(1): 317, 2018 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-30547803

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ciclina G2/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Gástricas/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Chlorocebus aethiops , Ciclina G2/biosíntesis , Ciclina G2/genética , Femenino , Genes Supresores de Tumor , Células HT29 , Células HeLa , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Nucleares/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Transfección , Vía de Señalización Wnt
12.
Biomed Res Int ; 2018: 6938482, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30420966

RESUMEN

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.


Asunto(s)
Ciclina G2/genética , Ciclina G2/metabolismo , Nefropatías Diabéticas/metabolismo , Animales , Línea Celular , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Noqueados , Vía de Señalización Wnt/genética
13.
Toxicol Sci ; 166(2): 370-381, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30169763

RESUMEN

Molecular mechanism and key factors responsible for cytotoxicity against mycotoxin deoxynivalenol (DON) from Fusarium pathogens are rarely elucidated. In this study, rapid increases of ROS were first observed in human gastric epithelial (GES-1) cells under DON exposure. Mitochondrial DNA damage, impaired respiratory chain, and decreased oxygen consumption rate (OCR) values, as well as G2/M cell cycle arrest and apoptosis, were also detected. Via combinatorial approaches of a large-scale microarray of differentially expressed genes, high content and RNAi analysis, a transcription factor of Forkhead box O3 (FOXO3a) was found with crucial functionalities, regulated some apoptotic genes associated with mitochondrial toxicity and cell death after activation by nuclear translocation. Namely, knockdown of FOXO3a decreased the cytotoxicity of DON to GES-1 cells. Moreover, knockdown of the FOXO ortholog DAF16 in Caenorhabditis elegans increased the resistance to DON-induced cytotoxicity. Simultaneously, the signaling pathway of ROS/JNK/FOXO3a of DON-induced cytotoxicity was newly proposed. In total, FOXO3a via ROS/JNK/FOXO3a plays a critical role to function as negative regulator associating with DON-induced cytotoxicity, with the potential extending to other substances.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Mitocondrias/efectos de los fármacos , Tricotecenos/toxicidad , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Caspasa 3/metabolismo , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Ciclina G2/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción Forkhead/metabolismo , Fusarium/química , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , MAP Quinasa Quinasa 4/metabolismo , Mitocondrias/metabolismo , Proteínas Nucleares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño , Especies Reactivas de Oxígeno , Transducción de Señal , Factor de Transcripción CHOP/metabolismo
14.
Oncol Rep ; 40(4): 2067-2078, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30106441

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias Colorrectales/patología , Ciclina G2/metabolismo , MicroARNs/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/cirugía , Ciclina G2/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Receptores Citoplasmáticos y Nucleares/genética , Tasa de Supervivencia , Células Tumorales Cultivadas
15.
Cell Physiol Biochem ; 44(5): 1741-1748, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29216623

RESUMEN

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.


Asunto(s)
Ciclina G2/metabolismo , Exosomas/metabolismo , MicroARNs/metabolismo , Regiones no Traducidas 3' , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Análisis por Conglomerados , Ciclina G2/antagonistas & inhibidores , Ciclina G2/genética , Resistencia a Antineoplásicos , Femenino , Humanos , Células MCF-7 , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Nuclear Pequeño/metabolismo , Alineación de Secuencia , Regulación hacia Arriba
16.
PLoS One ; 12(6): e0179906, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28640887

RESUMEN

Cyclin G2 (CCNG2) is an atypical cyclin that functions to inhibit cell cycle progression and is often dysregulated in human cancers. We have previously shown that cyclin G2 is highly unstable and can be degraded through the ubiquitin/proteasome pathway. Furthermore, cyclin G2 contains a PEST domain, which has been suggested to act as a signal for degradation by multiple proteases. In this study, we determined if calpains, a family of calcium-dependent proteases, are also involved in cyclin G2 degradation. The addition of calpain inhibitors or silencing of calpain expression by siRNAs strongly enhanced cyclin G2 levels. On the other hand, incubation of cell lysates with purified calpains or increasing the intracellular calcium concentration resulted in a decrease in cyclin G2 levels. Interestingly, the effect of calpain was found to be dependent on the phosphorylation of cyclin G2. Using a kinase inhibitor library, we found that Epidermal Growth Factor (EGF) Receptor is involved in cyclin G2 degradation and treatment with its ligand, EGF, induced cyclin G2 degradation. In addition, the presence of the PEST domain is necessary for calpain and EGF action. When the PEST domain was completely removed, calpain or EGF treatment failed to trigger degradation of cyclin G2. Taken together, these novel findings demonstrate that EGF-induced, calpain-mediated proteolysis contributes to the rapid destruction of cyclin G2 and that the PEST domain is critical for EGF/calpain actions.


Asunto(s)
Calpaína/metabolismo , Ciclina G2/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Proteolisis/efectos de los fármacos , Neoplasias del Cuello Uterino/patología , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Ciclina G2/química , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Femenino , Humanos , Fosforilación/efectos de los fármacos , Dominios Proteicos
17.
Sci Rep ; 6: 39091, 2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-27982046

RESUMEN

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.


Asunto(s)
Ciclina G1/genética , Ciclina G2/genética , Fibroblastos/citología , Neoplasias de Cabeza y Cuello/genética , Animales , Camptotecina/efectos adversos , Línea Celular Tumoral , Células Cultivadas , Quinasa de Punto de Control 2/metabolismo , Ciclina G1/metabolismo , Ciclina G2/metabolismo , Daño del ADN , Reparación del ADN , Regulación hacia Abajo , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Neoplasias de Cabeza y Cuello/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Fosforilación , Radiación Ionizante
18.
Cell Cycle ; 15(23): 3278-3295, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27753529

RESUMEN

Definition of cell cycle control proteins that modify tumor cell resistance to estrogen (E2) signaling antagonists could inform clinical choice for estrogen receptor positive (ER+) breast cancer (BC) therapy. Cyclin G2 (CycG2) is upregulated during cell cycle arrest responses to cellular stresses and growth inhibitory signals and its gene, CCNG2, is directly repressed by E2-bound ER complexes. Our previous studies showed that blockade of HER2, PI3K and mTOR signaling upregulates CycG2 expression in HER2+ BC cells, and that CycG2 overexpression induces cell cycle arrest. Moreover, insulin and insulin-like growth factor-1 (IGF-1) receptor signaling strongly represses CycG2. Here we show that blockade of ER-signaling in MCF7 and T47D BC cell lines enhances the expression and nuclear localization of CycG2. Knockdown of CycG2 attenuated the cell cycle arrest response of E2-depleted and fulvestrant treated MCF7 cells. These muted responses were accompanied by sustained inhibitory phosphorylation of retinoblastoma (RB) protein, expression of cyclin D1, phospho-activation of ERK1/2 and MEK1/2 and expression of cRaf. Our work indicates that CycG2 can form complexes with CDK10, a CDK linked to modulation of RAF/MEK/MAPK signaling and tamoxifen resistance. We determined that metformin upregulates CycG2 and potentiates fulvestrant-induced CycG2 expression and cell cycle arrest. CycG2 knockdown blunts the enhanced anti-proliferative effect of metformin on fulvestrant treated cells. Meta-analysis of BC tumor microarrays indicates that CCNG2 expression is low in aggressive, poor-prognosis BC and that high CCNG2 expression correlates with longer periods of patient survival. Together these findings indicate that CycG2 contributes to signaling networks that limit BC.


Asunto(s)
Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ciclina G2/metabolismo , Estradiol/análogos & derivados , Metformina/farmacología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Supervivencia sin Enfermedad , Estradiol/farmacología , Estrógenos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fulvestrant , Técnicas de Silenciamiento del Gen , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Recurrencia Local de Neoplasia/patología , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Interferencia de ARN , Receptores de Estrógenos/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Regulación hacia Arriba/efectos de los fármacos
19.
Mol Med Rep ; 13(1): 273-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26573378

RESUMEN

Colorectal cancer (CRC) is the third most common cancer type and the fourth leading cause of cancer­associated mortality worldwide. MicroRNA (miR)­1246 is involved in differentiation, invasion, metastasis and chemoresistance of certain types of tumor cells. CCNG2 encodes an unconventional cyclin homolog, cyclin G2 (CycG2), associated with growth inhibition, which correlated significantly with lymph node metastasis, clinical stage, histological grade and poor overall survival in numerous cancer types. To investigate the regulation of miR­1246 on CycG2 expression, and their effects on proliferation and metastasis of CRC, HCT­116 and LOVO cells were transfected with pre­miR­1246 anti­miR­1246 and their negative controls. It was demonstrated that the expression of miR­1246 was significantly increased in CRC tissues and cell lines, which was the opposite of CycG2. miR­1246 negatively regulated the expression of CycG2 in HCT­116 and LOVO CRC cells. CCNG2 is a direct target of miR­1246 in CRC cells. Overexpression of miR­1246 induced cell proliferation, migration and invasion, while knockdown of miR­1246 inhibited proliferation, migration and invasion in the CRC cells. Upregulation of miR­1246 mediated the malignant progression of CRC and is partly attributed to the downregulation of the expression of CycG2. Consequently, these findings provided a molecular basis for the role of miR­1246/CCNG2 in the progression of human CRC and suggested a novel target for the treatment of CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ciclina G2/metabolismo , MicroARNs/metabolismo , Anciano , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Cell Microbiol ; 18(1): 46-65, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26149128

RESUMEN

Microbial pathogens have developed several mechanisms to modulate and interfere with host cell cycle progression. In this study, we analysed the effect of the human pathogen Neisseria meningitidis on cell cycle in a brain endothelial cell line as well as in primary brain endothelial cells. We found that N. Meningitidis causes an accumulation of cells in the S phase early at 3 and at 24 h post-infection that was paralleled by a decrease of cells in G2/M phase. Importantly, the outer membrane proteins of the colony opacity-associated (Opa) protein family as well as the Opc protein proved to trigger the accumulation of cells in the S phase. A focused cell cycle reverse transcription quantitative polymerase chain reaction-based array and integrated network analysis revealed changes in the abundance of several cell cycle regulatory mRNAs, including the cell cycle inhibitors p21(WAF1/CIP1) and cyclin G2. These alterations were reflected in changes in protein expression levels and/or relocalization in N. meningitidis-infected cells. Moreover, an increase in p21(WAF1/CIP1) expression was found to be p53 independent. Genetic ablation of p21(WAF1/CIP1) and cyclin G2 abrogated N. meningitidis-induced S phase accumulation. Finally, by measuring the levels of the biomarker 8-hydroxydeoxyguanosine and phosphorylation of the histone variant H2AX, we provide evidence that N. meningitidis induces oxidative DNA damage in infected cells.


Asunto(s)
Puntos de Control del Ciclo Celular , Ciclina G2/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Endoteliales/microbiología , Interacciones Huésped-Patógeno , Neisseria meningitidis/patogenicidad , Fase S , Proteínas de la Membrana Bacteriana Externa/metabolismo , Encéfalo , Células Cultivadas , Células Endoteliales/fisiología , Perfilación de la Expresión Génica , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...