Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Pharmacol Res ; 180: 106242, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35513227

RESUMO

Cancer progression is enhanced through cell proliferation, with the crucial role of the transducer and transmembrane -signal regulator (GNG12) bringing it to the fore. Dysregulation of cancer cell metabolism, evasion of the immune system, cell cycle, apoptosis, and chemoresistance result from inconsistent initiation of the NF-kB signaling pathway. We excerpt from previous studies that overactivation of the canonic NF-kB cascade occurs in varieties of tumor cells, which results in the growth of lymphovascular invasion, as well as neural invasion. Recently, research has adduced that a particular G protein- coupled receptor (GNG12) is silently involved in the activation of the NF-kB signal, which supports the evasion of cancer immunity and in turn activates cancer proliferation, angiogenesis, and immunotherapeutic resistance. While the likely impact of GNG12 in relation to the progression of tumors is being established, there is insufficient knowledge regarding the functions and mechanisms of GNG12 in cancer immunity. Furthermore, the cancer-associated role as well as the clinical correlation of GNG12 have long been unknown; thus, their identification is more likely to pave the path for a novel regime of tumor suppression. In this study, we established the silent role of GNG12 in activating NF-kB genes and the synergism between NF-kB and PD-L1 expression. Captivatingly, we reported that silencing GNG12 gene downregulates the transcription of PD-L1 gene. We therefore suggested that GNG12 is a risk factor for several cancers, and a possible target for immunotherapy.


Assuntos
Antígeno B7-H1 , Subunidades gama da Proteína de Ligação ao GTP , Neoplasias , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Proliferação de Células , Subunidades gama da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/imunologia , Humanos , NF-kappa B/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Transdução de Sinais
2.
J Clin Lab Anal ; 36(3): e24207, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35060646

RESUMO

OBJECTIVE: Glioma is the most common, rapidly progressing, lethal brain tumor. However, underlying mechanisms behind its abnormal progression remain largely unknown. This study aimed to investigate mechanism of action and effects of the hsa_circ_0000285 on glioma progression. METHODS: RT-qPCR was utilized to study RNA expression in glioma tissues and cell lines. The effects of hsa_circ_0000285 on glioma progression were studied by measuring cell proliferation and migration, apoptosis, tumor volume and weight in both glioma cells and xenograft glioma mice. The features of hsa_circ_0000285 were identified using chromatin fractionation and RNase digestion. Its mechanism of action was analyzed using bioinformatics, RNA-binding protein immunoprecipitation, and luciferase reporter assay. RESULTS: We found glioma tissues and cell lines were overexpressing hsa_circ_0000285. While hsa_circ_0000285 promoted cell proliferation and migration, it inhibited apoptosis in vitro. It also increased tumor volume and weight in vivo. Using bioinformatic analysis and verification experiments for studying its mechanisms, we confirmed that hsa_circ_0000285 sponged miR-599, which negatively regulated GNG12 by binding to its mRNA. CONCLUSION: Hsa_circ_0000285 is overexpressed in the glioma and promotes its progression by directly regulating the miR-599/GNG12 axis. This novel mechanism, therefore, shows that the hsa_circ_0000285/miR-599/GNG12 axis may be a promising therapeutic target for glioma treatment.


Assuntos
Glioma , MicroRNAs , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Circular/genética
3.
Int J Mol Sci ; 21(9)2020 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-32365959

RESUMO

We hypothesized that DNA methylation patterns may contribute to the development of active pulmonary tuberculosis (TB). Illumina's DNA methylation 450 K assay was used to identify differentially methylated loci (DML) in a discovery cohort of 12 active pulmonary TB patients and 6 healthy subjects (HS). DNA methylation levels were validated in an independent cohort of 64 TB patients and 24 HS. Microarray analysis identified 1028 DMLs in TB patients versus HS, and 3747 DMLs in TB patients after versus before anti-TB treatment, while autophagy was the most enriched signaling pathway. In the validation cohort, PARP9 and miR505 genes were hypomethylated in the TB patients versus HS, while RASGRP4 and GNG12 genes were hypermethylated, with the former two further hypomethylated in those with delayed sputum conversion, systemic symptoms, or far advanced lesions. MRPS18B and RPTOR genes were hypomethylated in TB patients with pleural involvement. RASGRP4 gene hypermethylation and RPTOR gene down-regulation were associated with high mycobacterial burden. TB patients with WIPI2/GNG12 hypermethylation or MRPS18B/FOXO3 hypomethylation had lower one-year survival. In vitro ESAT6 and CFP10 stimuli of THP-1 cells resulted in DNA de-methylation changes of the PARP9, RASGRP4, WIPI2, and FOXO3 genes. In conclusions, aberrant DNA methylation over the PARP9/miR505/RASGRP4/GNG12 genes may contribute to the development of active pulmonary TB disease and its clinical phenotypes, while aberrant DNA methylation over the WIPI2/GNG12/MARPS18B/FOXO3 genes may constitute a determinant of long-term outcomes.


Assuntos
Metilação de DNA/fisiologia , Regiões Promotoras Genéticas/genética , Tuberculose Pulmonar/genética , Estudos de Coortes , Metilação de DNA/genética , Proteína Forkhead Box O3/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Proteínas de Ligação a Fosfato/genética , Poli(ADP-Ribose) Polimerases/genética , Proteína Regulatória Associada a mTOR/genética , Fatores ras de Troca de Nucleotídeo Guanina/genética
4.
Cancer Biomark ; 40(1): 61-78, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38277283

RESUMO

BACKGROUND: Breast cancer is the most commonly occurring cancer worldwide and is the main cause of death from cancer in women. Novel biomarkers are highly warranted for this disease. OBJECTIVE: Evaluation of novel long non-coding RNAs biomarkers for breast cancer. METHODS: The study comprised the analysis of the expression of 71 candidate lncRNAs via screening, six of which (four underexpressed, two overexpressed) were validated and analyzed by qPCR in tumor tissues associated with NST breast carcinomas, compared with the benign samples and with respect to their clinicopathological characteristics. RESULTS: The results indicated the tumor suppressor roles of PTENP1, GNG12-AS1, MEG3 and MAGI2-AS3. Low levels of both PTENP1 and GNG12-AS1 were associated with worsened progression-free and overall survival rates. The reduced expression of GNG12-AS1 was linked to the advanced stage. A higher grade was associated with the lower expression of PTENP1, GNG12-AS1 and MAGI2-AS3. Reduced levels of both MEG3 and PTENP1 were linked to Ki-67 positivity. The NRSN2-AS1 and UCA1 lncRNAs were overexpressed; higher levels of UCA1 were associated with multifocality. CONCLUSIONS: The results suggest that the investigated lncRNAs may play important roles in breast cancer and comprise a potential factor that should be further evaluated in clinical studies.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , Feminino , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Pessoa de Meia-Idade , Regulação Neoplásica da Expressão Gênica , Adulto , Prognóstico , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Guanilato Quinases/genética , Guanilato Quinases/metabolismo , Idoso , Gradação de Tumores , Genes Supressores de Tumor
5.
Am J Cancer Res ; 13(12): 5981-5995, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38187053

RESUMO

Porphyromonas gingivalis (P. gingivalis), a Gram-negative oral anaerobe, was demonstrated to facilitate colonization and progression in colonic tumor, while the underlying mechanism still remains to be clarified. Here, we identified the proteome profile changed by P. gingivalis infection in HCT116 cells through label-free quantitative proteomics, and found that deubiquitinase UCHL3 was a key protein that response for P. gingivalis infection. By CCK8, colony formation, wound healing assays, and in vivo subcutaneous tumor mouse moudle, we proved that P. gingivalis could promote the proliferation and migration of colon cancer, while the process was inhibited by UCHL3 knock down. Through IP-MS, we identified GNG12 as the UCHL3 interacting protein. The protein level of GNG12 was significantly reduced when knock out UCHL3. Thus we propose that GNG12 is a substrate protein of UCHL3. Furthermore, we demonstrated that overexpression of GNG12 could restore the tumor inhibition effect caused by UCHL3 knock down, and UCHL3-GNG12 axis promote colon cancer progression via the NF-κB signal pathway. Collectively, this study unveiled that P. gingivalis infection up-regulated UCHL3 and stabilized its substrate protein GNG12 to activate the NF-κB signal pathway to promote colon cancer progression. Our study indicate that UCHL3 is a potential biomarker and therapeutic target for colon cancer which infected with P. gingivalis.

6.
Hematology ; 28(1): 2182169, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36861936

RESUMO

OBJECTIVE: To identify biomarkers that can predict the recurrence of the central nervous system (CNS) in children with acute lymphoblastic leukemia (ALL). MATERIALS AND METHODS: The transcriptome and clinical data of ALL in children were downloaded from the TARGET database. Transcriptome data were analyzed by bioinformatics method to identify core (hub) genes and establish a risk assessment model. Univariate Cox analysis was performed on each clinical data, and multivariate Cox regression analysis was performed on the obtained results and risk score. The children ALL phase I samples from TARGET database were used for validation. RESULTS: Univariate multivariate Cox analysis of 10 hub genes identified showed that PPARG (HR = 0.78, 95%CI = 0.67-0.91, p = 0.007), CD19 (HR = 1.15, 95%CI = 1.05-1.26, p = 0.003) and GNG12 (HR = 1.25, 95%CI = 1.04-1.51, p = 0.017) had statistical differences. The risk score was statistically significant in univariate (HR = 3.06, 95%CI = 1.30-7.19, p = 0.011) and multivariate (HR = 1.81, 95%CI = 1.16-2.32, p = 0.046) Cox regression analysis. The survival analysis results of the high and low-risk groups were different when the validation dataset was substituted into the model (p = 0.018). Then, we constructed a Nomogram which had a concordance index of survival prediction of 0.791(95%CI= 0.779-0.803). In addition, the CNS involvement grading status at first diagnosis CNS3 vs. CNS1 (HR = 5.74, 95%CI = 2.01-16.4, p = 0.001), T cell vs B cell (HR = 1.63, 95% CI = 1.06-2.49, p = 0.026) were also statistically significant. CONCLUSIONS: PPARG, GNG12, and CD19 may be predictors of CNS relapse in childhood ALL.


Assuntos
PPAR gama , Leucemia-Linfoma Linfoblástico de Células Precursoras , Criança , Humanos , Proteínas Adaptadoras de Transdução de Sinal , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Linfócitos B , Biologia Computacional
7.
J Mol Neurosci ; 72(2): 441-450, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35083664

RESUMO

Glioma is one of the most aggressive malignancies and has a poor survival rate. G protein subunit gamma 12 (GNG12), a member of G protein family, has been reported to participate in cancer disorders. However, the role and functional mechanism of GNG12 in glioma are not fully understood. The expression of GNG12 mRNA and miR-876-5p was measured by qRT-PCR. The level of GNG12 protein was measured by western blot. Cell proliferation and cell migration were monitored by CCK-8 assay and wound healing assay. The role of GNG12 on tumorigenicity in vivo was determined by animal models. The interaction between GNG12 and miR-876-5p was validated by dual-luciferase reporter experiment. The phosphorylation levels of PI3K and AKT were monitored by western blot. The upregulated expression of GNG12 was identified in tumor tissues and cells of glioma. GNG12 knockdown inhibited glioma cell growth and migration, and slowed tumor development in vivo. Besides, GNG12 knockdown weakened the phosphorylation levels of PI3K and AKT. GNG12 was verified to be a target of miR-876-5p whose enrichment suppressed the expression and function of GNG12. MiR-876-5p repressed glioma cell proliferation, migration, and the activity of PI3K/AKT signaling by targeting GNG12. MiR-876-5p-targeted GNG12 contributes to the malignant development of glioma by increasing the PI3K/AKT signaling activity.


Assuntos
Subunidades gama da Proteína de Ligação ao GTP , Glioma , MicroRNAs , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Glioma/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
8.
Front Immunol ; 12: 758845, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691083

RESUMO

Background: G protein subunit gamma 12 (GNG12) is observed in some types of cancer, but its role in osteosarcoma is unknown. This study hypothesized that GNG12 may be a potential biomarker and therapeutic target. We aimed to identify an association between GNG12 and osteosarcoma based on the Gene Expression Omnibus and the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) databases. Methods: Osteosarcoma samples in GSE42352 and TARGET database were selected as the test cohorts. As the external validation cohort, 78 osteosarcoma specimens from The Second Affiliated Hospital of Nanchang University were collected. Patients with osteosarcoma were divided into high and low GNG12 mRNA-expression groups; differentially expressed genes were identified as GNG12-related genes. The biological function of GNG12 was annotated using Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, gene set enrichment analysis, and immune infiltration analysis. Gene expression correlation analysis and competing endogenous RNA regulatory network construction were used to determine potential biological regulatory relationships of GNG12. Overall survival, Kaplan-Meier analysis, and log-rank tests were calculated to determine GNG12 reliability in predicting survival prognosis. Results: GNG12 expression decreased in osteosarcoma samples. GNG12 was a highly effective biomarker for osteosarcoma [area under the receiver operating characteristic (ROC) curve (AUC) = 0.920], and the results of our Kaplan-Meier analysis indicated that overall survival and progression-free survival differed significantly between low and high GNG-expression group (p < 0.05). Functional analyses indicated that GNG12 may promote osteosarcoma through regulating the endoplasmic reticulum. Expression correlation analysis and competing endogenous RNA network construction showed that HOTTIP/miR-27a-3p may regulate GNG12 expression. Furthermore, the subunit suppresses adaptive immunity via inhibiting M1 and M2 macrophage infiltration. GNG12 was inhibited in metastatic osteosarcoma compared with non-metastatic osteosarcoma, and its expression predicted survival of patients (1, 3, and 5-year AUCs were 0.961, 0.826, and 0.808, respectively). Conclusion: This study identified GNG12 as a potential biomarker for osteosarcoma prognosis, highlighting its potential as an immunotherapy target.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Ósseas/imunologia , Subunidades gama da Proteína de Ligação ao GTP/genética , Osteossarcoma/imunologia , Biomarcadores Tumorais/imunologia , Neoplasias Ósseas/patologia , Subunidades gama da Proteína de Ligação ao GTP/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Mastócitos/imunologia , Mastócitos/patologia , Osteossarcoma/patologia , Microambiente Tumoral/imunologia
9.
FEBS Open Bio ; 10(2): 278-287, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31898405

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive solid tumors in the digestive system. A greater understanding of the pathogenesis of PDAC may facilitate the search for new therapeutic targets. Guanine nucleotide-binding protein subunit gamma-12 (GNG12) belongs to the G protein family and participates in the modulation of the inflammatory signaling cascade. However, the cancer-related function and clinical relevance of GNG12 in PDAC have not previously been reported. Here, we investigated the clinical significance of GNG12 in PDAC using the Oncomine web tool, the gene expression profiling interactive analysis tool and tissue microarray (TMA). GNG12 expression was observed to be higher in PDAC patient specimens than in nontumor pancreatic tissues, and high expression of GNG12 was associated with poor prognosis. We subsequently show that GNG12 promotes pancreatic cancer cell growth in vivo and in vitro, as evaluated using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt assays, colony formation assays and a xenograft mouse model. Furthermore, our results suggest that GNG12 activates nuclear factor-κB signaling and modulates the immune response. Collectively, our findings suggest that GNG12 may be suitable as a new prognosis-related biomarker and a promising target for treatment of pancreatic cancer.


Assuntos
Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Apoptose/genética , Antígeno B7-H1/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Bases de Dados Genéticas , Subunidades gama da Proteína de Ligação ao GTP/genética , Expressão Gênica , Humanos , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Transdução de Sinais/genética , Transcriptoma , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
10.
Biosci Rep ; 40(8)2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32735016

RESUMO

BACKGROUND: Long non-coding RNAs (lncRNAs) are increasingly being regarded as regulators of glioma development. Notably, some studies report that GNG12-AS1 plays important functions and molecular mechanism in breast cancer, but there are no existing studies in glioma. OBJECTIVE: To analyze the biological functions and potential mechanisms of GNG12-AS1 in glioma. METHODS: We detected the expression of GNG12-AS1 in glioma tissues through analyzing TCGA data as well as our clinical samples. We then evaluated cell proliferation through MTT assay and colony formation and cell migration by transwell assay, wound healing assay and single cell tracking assay. After, we analyzed the effects of the AKT/GSK-3ß/ß-catenin through Western blotting and utilized the ß-catenin agonist SKL2001 for the rescue experiment. RESULTS: GNG12-AS1 was highly expressed in glioma tissues. The silence of GNG12-AS1 inhibited the proliferation, migration and epithelial-mesenchymal transition of glioma cells, and reduced the activity of the AKT/GSK-3ß/ß-catenin pathway. Notably, SKL2001 could reverse cell migration as well as ß-catenin expression in glioma cells with lower GNG12-AS1 expression. CONCLUSIONS: GNG12-AS1 regulates proliferation and migration of glioma cells through the AKT/GSK-3ß/ß-catenin signaling and can perhaps be a new target for the treatment of glioma.


Assuntos
Neoplasias Encefálicas/enzimologia , Glioma/enzimologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , beta Catenina/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/patologia , Humanos , Invasividade Neoplásica , Interferência de RNA , RNA Longo não Codificante/genética , Via de Sinalização Wnt
11.
Biochim Biophys Acta Proteins Proteom ; 1866(11): 1092-1101, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30282607

RESUMO

In cow mammary epithelial cells (CMECs), cell growth and casein synthesis are regulated by amino acids (AAs), and lysosomes are important organelles in this regulatory process, but the mechanisms remain unclear. Herein, lysosomal membrane proteins (LMPs) in CMECs in the presence (Leu+) and absence (Leu-) of leucine were quantitatively analysed using Sequential Windowed Acquisition of All Theoretical Fragment Ion (SWATH) mass spectrometry. In identified LMPs, Guanine nucleotide-binding protein subunit gamma-12 (GNG12) was a markedly up-regulated protein in Leu+ group. CMECs were treated with Leu+ or Leu-, expression and lysosomal localization of GNG12 were decreased in response to Leu absence. Overexpressing or inhibiting GNG12 demonstrated that cell growth, casein synthesis and activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway were all up-regulated by GNG12. Cell growth, casein synthesis and mTORC1 signaling pathway were decreased in response to Leu absence, but these decreases were partially restored by GNG12 overexpression, and those effects were partially reversed by inhibiting GNG12. Co-immunoprecipitation analysis showed that GNG12 activates the mTORC1 pathway via interaction with Ragulator. Taken together, these results suggest that GNG12 is a positive regulator of the Leu-mediated mTORC1 signaling pathway in CMECs that promotes cell growth and casein synthesis.


Assuntos
Proliferação de Células , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Leucina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Caseínas/biossíntese , Bovinos , Células Cultivadas , Células Epiteliais/metabolismo , Feminino , Subunidades gama da Proteína de Ligação ao GTP/genética , Proteínas de Membrana Lisossomal/metabolismo , Lisossomos/metabolismo , Glândulas Mamárias Animais/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteômica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA