Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 23(12)2022 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-35743282

RESUMO

TRIM28 is a scaffold protein that interacts with DNA-binding proteins and recruits corepressor complexes to cause gene silencing. TRIM28 contributes to physiological functions such as cell growth and differentiation. In the chronic myeloid leukemia cell line K562, we edited TRIM28 using CRISPR/Cas9 technology, and the complete and partial knockout (KO) cell clones were obtained and confirmed using quantitative droplet digital PCR (ddPCR) technology. The amplicon sequencing demonstrated no off-target effects in our gene editing experiments. The TRIM28 KO cells grew slowly and appeared red, seeming to have a tendency towards erythroid differentiation. To understand how TRIM28 controls K562 cell proliferation and differentiation, transcriptome profiling analysis was performed in wild-type and KO cells to identify TRIM28-regulated genes. Some of the RNAs that encode the proteins regulating the cell cycle were increased (such as p21) or decreased (such as cyclin D2) in TRIM28 KO cell clones; a tumor marker, the MAGE (melanoma antigen) family, which is involved in cell proliferation was reduced. Moreover, we found that knockout of TRIM28 can induce miR-874 expression to downregulate MAGEC2 mRNA via post-transcriptional regulation. The embryonic epsilon-globin gene was significantly increased in TRIM28 KO cell clones through the downregulation of transcription repressor SOX6. Taken together, we provide evidence to demonstrate the regulatory network of TRIM28-mediated cell growth and erythroid differentiation in K562 leukemia cells.


Assuntos
Edição de Genes , MicroRNAs , Sistemas CRISPR-Cas , Proliferação de Células/genética , Expressão Gênica , Subunidades de Hemoglobina/genética , Subunidades de Hemoglobina/metabolismo , Humanos , Células K562 , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo
2.
Gastroenterology ; 153(4): 1107-1119.e10, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28648905

RESUMO

BACKGROUND & AIMS: Ligand binding to inhibitory receptors on immune cells, such as programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA4), down-regulates the T-cell-mediated immune response (called immune checkpoints). Antibodies that block these receptors increase antitumor immunity in patients with melanoma, non-small-cell lung cancer, and renal cell cancer. Tumor-infiltrating CD4+ and CD8+ T cells in patients with hepatocellular carcinoma (HCC) have been found to be functionally compromised. We analyzed HCC samples from patients to determine if these inhibitory pathways prevent T-cell responses in HCCs and to find ways to restore their antitumor functions. METHODS: We collected HCC samples from 59 patients who underwent surgical resection from November 2013 through May 2017, along with tumor-free liver tissues (control tissues) and peripheral blood samples. We isolated tumor-infiltrating lymphocytes (TIL) and intra-hepatic lymphocytes. We used flow cytometry to quantify expression of the inhibitory receptors PD-1, hepatitis A virus cellular receptor 2 (TIM3), lymphocyte activating 3 (LAG3), and CTLA4 on CD8+ and CD4+ T cells from tumor, control tissue, and blood; we studied the effects of antibodies that block these pathways in T-cell activation assays. RESULTS: Expression of PD-1, TIM3, LAG3, and CTLA4 was significantly higher on CD8+ and CD4+ T cells isolated from HCC tissue than control tissue or blood. Dendritic cells, monocytes, and B cells in HCC tumors expressed ligands for these receptors. Expression of PD-1, TIM3, and LAG3 was higher on tumor-associated antigen (TAA)-specific CD8+ TIL, compared with other CD8+ TIL. Compared with TIL that did not express these inhibitory receptors, CD8+ and CD4+ TIL that did express these receptors had higher levels of markers of activation, but similar or decreased levels of granzyme B and effector cytokines. Antibodies against CD274 (PD-ligand1 [PD-L1]), TIM3, or LAG3 increased proliferation of CD8+ and CD4+ TIL and cytokine production in response to stimulation with polyclonal antigens or TAA. Importantly, combining antibody against PD-L1 with antibodies against TIM3, LAG3, or CTLA4 further increased TIL functions. CONCLUSIONS: The immune checkpoint inhibitory molecules PD-1, TIM3, and LAG3 are up-regulated on TAA-specific T cells isolated from human HCC tissues, compared with T cells from tumor-free liver tissues or blood. Antibodies against PD-L1, TIM3, or LAG3 restore responses of HCC-derived T cells to tumor antigens, and combinations of the antibodies have additive effects. Strategies to block PD-L1, TIM3, and LAG3 might be developed for treatment of primary liver cancer.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Antígenos CD , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Receptor Celular 2 do Vírus da Hepatite A/antagonistas & inibidores , Imunoterapia/métodos , Neoplasias Hepáticas/tratamento farmacológico , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Antígenos CD/imunologia , Antígenos CD/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Antígeno CTLA-4/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Ativação Linfocitária/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Evasão Tumoral/efeitos dos fármacos , Microambiente Tumoral , Regulação para Cima , Proteína do Gene 3 de Ativação de Linfócitos
3.
BMC Cancer ; 18(1): 971, 2018 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-30309319

RESUMO

BACKGROUND: Cancer/testis antigen MAGEC2 (also known as HCA587) is highly expressed in a wide variety of tumors and plays an active role in promoting growth and metastasis of tumor cells. However, little is known for the regulation of MAGEC2 expression in cancer cells. METHODS: Western blotting and quantitative RT-PCR were performed to analyze MAGEC2 expression. Co-immunoprecipitation assay was applied for detecting the endogenous interaction of MAGEC2 and TRIM28 in tumor cells. Overexpression and knockdown assays were used to examine the effects of TRIM28 on the expression of MAGEC2 protein. Immunohistochemistry (IHC) staining was performed in hepatocellular carcinoma patients to evaluate the association between the expression of MAGEC2 and TRIM28. Proteasome inhibitors MG132 or PS-341 and lysosome inhibitor Chloroquine (CQ) were used to inhibit proteasomal or lysosomal-mediated protein degradation respectively. RESULTS: We demonstrate that MAGEC2 interacts with TRIM28 in melanoma cells and MAGEC2 expression in tumor cells depends on the expression of TRIM28. The expression level of MAGEC2 protein was significantly reduced when TRIM28 was depleted in tumor cells, and no changes were observed in MAGEC2 mRNA level. Furthermore, expression levels of MAGEC2 and TRIM28 are positively correlated in MAGEC2-positive human hepatocellular carcinoma tissues (p = 0.0011). Mechanistic studies indicate that the regulatory role of TRIM28 on MAGEC2 protein expression in tumor cells depends on proteasome-mediated pathway. CONCLUSIONS: Our findings show that TRIM28 is necessary for MAGEC2 expression in cancer cells, and TRIM28 may serve as a new potential target for immunotherapy of cancer.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Células A549 , Antígenos de Neoplasias/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Melanoma/genética , Melanoma/metabolismo , Proteínas de Neoplasias/genética , Neoplasias/genética , Ligação Proteica , Proteína 28 com Motivo Tripartido/genética
4.
Cancer Sci ; 107(12): 1888-1897, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27636589

RESUMO

Cancer/testis antigen MAGEC2, a member of the type I melanoma-associated antigen family, is expressed in a wide variety of cancer types but not in normal somatic cells. MAGEC2 has long been recognized as a tumor-specific target, however, its functions remain largely unknown. In this study, we established MAGEC2-knockout A375 melanoma cell lines using the CRISPR/Cas9 system. Seven clonal cell lines were generated by using four single guide RNAs targeting the coding region of the MAGEC2 gene, which produced indels that abolished MAGEC2 protein expression. To identify the differentially expressed protein profiles associated with MAGEC2 loss, isobaric tag for relative quantitation-based comparative proteomics experiments were carried out on the MAGEC2-knockcout and control A375 cells. Mining of the proteomics data identified a total 224 (61.6% upregulated and 38.4% downregulated) proteins to be significantly altered in expression level in MAGEC2-knockcout cells. Ingenuity Pathway Analysis indicated that the significantly altered proteins were involved in critical neoplasia-related biological functions such as cell death, proliferation, and movement. Gene ontology analysis identified "apoptosis signaling" as the top-most upregulated pathway associated with MAGEC2 loss. We showed that knockout or knockdown of the MAGEC2 gene sensitized melanoma cells to tumor necrosis factor-α-induced apoptosis. Interestingly, actin-based motility by Rho and RhoA signaling, known to promote cell migration, were also identified as the top downregulated pathways in MAGEC2-knockout A375 cells. In short, our study provides a suitable cell model for exploring the biological functions of MAGEC2 in malignant cells, and sheds light on the molecular pathway by which MAGEC2 promotes tumor development.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Modelos Animais de Doenças , Deleção de Genes , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Redes Reguladoras de Genes , Marcação de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Knockout , Mapas de Interação de Proteínas , Proteoma , Proteômica/métodos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
5.
Br J Haematol ; 171(5): 752-62, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26456863

RESUMO

Cancer-testis antigens belonging to the MAGE family of genes, such as MAGEC2, are commonly and specifically expressed in Multiple Myeloma (MM) and are associated with a more aggressive clinical course and chemotherapy resistance. MAGEC2 is thought to be an excellent candidate for cancer immunotherapy; however, the biological role of MAGEC2 in MM has remained unclear. We investigated the biological role of MAGEC2 in myeloma cells determining the effect of MAGEC2 knockdown on proliferation and apoptosis. Loss of MAGEC2 resulted in reduced proliferation, viability, and anchorage-independent growth of myeloma cells irrespective of the functional status of TP53 (p53). The anti-proliferative effect of MAGEC2 silencing was due to a decrease of cells in the S phase, cell cycle delay at both G0/G1 and/or G2/M, and an increase in the sub-G0/G1 diploid population related to apoptotic cell death. Importantly, overexpression of short hairpin (sh)RNA-refractory MAGEC2 rescued the anti-proliferative effect of mRNA knockdown and protected cells from apoptotic cell death. Our findings support a TP53-independent role of MAGEC2 in promoting the survival of myeloma cells suggesting that MAGEC2-specific immunotherapies have the potential to eradicate the most malignant cells within the myeloma tumour bulk leading to durable clinical responses.


Assuntos
Antígenos de Neoplasias/fisiologia , Apoptose/fisiologia , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/fisiologia , Antígenos de Neoplasias/genética , Ciclo Celular/genética , Crescimento Celular , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/genética , Técnicas de Silenciamento de Genes , Humanos , Mutação de Sentido Incorreto/genética , Proteínas de Neoplasias/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Proteína Supressora de Tumor p53/fisiologia
6.
Cartilage ; 13(3): 19476035221121790, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36068934

RESUMO

OBJECTIVE: It is unknown why some athletes develop chondromalacia and others do not, even when accounting for similar workloads between individuals. Genetic differences between individuals may be a contributing factor. The purpose of this work was to screen the entire genome for genetic markers associated with chondromalacia. DESIGN: Genome-wide association (GWA) analyses were performed utilizing data from the Kaiser Permanente Research Board (KPRB) and the UK Biobank. Chondromalacia cases were identified based on electronic health records from KPRB and UK Biobank. GWA analyses from both cohorts were tested for chondromalacia using a logistic regression model adjusting for sex, height, weight, age of enrollment, and race/ethnicity using allele counts for single-nucleotide polymorphisms (SNPs). The data from the 2 GWA studies (KPRB and UK Biobank) were combined in a meta-analysis. RESULTS: There were a total of 3,872 combined cases of chondromalacia from the KPRB and the UK Biobank cohorts. Genome-wide significant associations with chondromalacia were found for rs144449054 in the ARHGAP15 gene (OR = 3.70 [2.32-5.90]; P = 1.4 × 10-8) and rs188900564 in the MAGEC2 (OR = 2.07 [1.61-2.65]; P = 3.7 × 10-9). CONCLUSIONS: Genetic markers in ARHGAP15 and MAGEC2 appear to be associated with chondromalacia and are potential risk factors that deserve further validation regarding molecular mechanisms.


Assuntos
Doenças das Cartilagens , Estudo de Associação Genômica Ampla , Marcadores Genéticos/genética , Humanos , Polimorfismo de Nucleotídeo Único/genética
7.
Front Endocrinol (Lausanne) ; 13: 816598, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35355564

RESUMO

The expression of the melanoma/cancer-testis antigen MAGEC2/CT10 is restricted to germline cells, but like most cancer-testis antigens, it is frequently upregulated in advanced breast tumors and other malignant tumors. However, the physiological cues that trigger the expression of this gene during malignancy remain unknown. Given that malignant breast cancer is often associated with skeletal metastasis and co-morbidities such as cancer-induced hypercalcemia, we evaluated the effect of high Ca2+ on the calcium-sensing receptor (CaSR) and potential mechanisms underlying the survival of triple-negative breast cancer (TNBC) cells at high Ca2+. We show that chronic exposure of TNBC cells to high Ca2+ decreased the sensitivity of CaSR to Ca2+ but stimulated tumor cell growth and migration. Furthermore, high extracellular Ca2+ also stimulated the expression of early response genes such as FOS/FOSB and a unique set of genes associated with malignant tumors, including MAGEC2. We further show that the MAGEC2 proximal promoter is Ca2+ inducible and that FOS/FOSB binds to this promoter in a Ca2+- dependent manner. Finally, downregulation of MAGEC2 strongly inhibited the growth of TNBC cells in vitro. These data suggest for the first time that MAGEC2 is a high Ca2+ inducible gene and that aberrant expression of MAGEC2 in malignant TNBC tissues is at least in part mediated by an increase in circulating Ca2+via the AP-1 transcription factor.


Assuntos
Hipercalcemia , Neoplasias de Mama Triplo Negativas , Antígenos de Neoplasias , Cálcio , Linhagem Celular Tumoral , Humanos , Masculino , Proteínas de Neoplasias , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
8.
Onco Targets Ther ; 12: 7843-7855, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31576142

RESUMO

PURPOSE: Although MAGEC2 was first cloned from a human hepatocellular carcinoma (HCC) cDNA library by serum screening, the detailed attributes of MAGEC2 in HCC have rarely been elucidated. PATIENTS AND METHODS: In this study, The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases were consulted to analyse the expression of MAGEC2 mRNA in liver cancer. Immunohistochemistry (IHC) analysis was performed to detect MAGEC2 expression in HCC, and the relationship between MAGEC2 expression and the clinicopathological characteristics of HCC patients was evaluated. Then, we employed the short hairpin (sh)RNA-mediated knockdown of MAGEC2 in HCC cell lines to explore the function of MAGEC2 in HCC development. Finally, the expression of epithelial-mesenchymal transition (EMT) markers in HCC xenografts and clinical samples was investigated. RESULTS: The results showed a remarkably higher level of MAGEC2 expression in HCC tissues than in noncancerous tissues, and MAGEC2 expression could be used as an independent prognostic factor for overall survival in HCC. Moreover, sh-MAGEC2 inhibited a series of HCC malignant behaviours both in vitro and in vivo. Finally, decreased MAGEC2 expression and low levels of EMT markers were detected in sh-MAGEC2 xenografts, while increased MAGEC2 expression and high levels of EMT markers were observed in invasive and metastatic HCC samples. CONCLUSION: Taken together, our data imply that MAGEC2 is a novel prognostic marker for HCC and that MAGEC2 significantly promotes HCC tumourigenesis by inducing EMT. Targeting MAGEC2 may provide a promising therapeutic strategy for HCC treatment.

9.
Technol Cancer Res Treat ; 17: 1533033818797587, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30198403

RESUMO

OBJECTIVE: To investigate the role of MAGE family member C2 in angiogenesis and epithelial-mesenchymal transition of non-small cell lung carcinoma. METHODS: The Cancer Genome Atlas data set was analyzed to filter the highly expressed gene melanoma antigen family C2 in non-small cell lung carcinoma. Quantitative reverse transcription-polymerase chain reaction was performed to verify the overexpression of melanoma antigen family C2 in non-small cell lung carcinoma cell lines. Melanoma antigen family C2 complementary DNA and short hairpin RNA (shRNA) were transfected into SK-MES-1 cells to regulate melanoma antigen family C2 expression. Cell Counting Kit-8 assay, flow cytometry, wound healing assay, and Transwell assay were performed to investigate the effect of melanoma antigen family C2 on proliferation, apoptosis, migration, and invasion of SK-MES-1 cell line. Western blot was used to detect the expression of epithelial-mesenchymal transition markers. Enzyme-linked immunosorbent assay was performed to investigate the secretion of vascular endothelial growth factor, and tube formation assay was conducted to explore the effect of melanoma antigen family C2 on angiogenesis ability of the tumor. Tumor xenograft on nude mice and immunohistochemical/hematoxylin and eosin staining were also performed to detect the influence of melanoma antigen family C2 on growth and metastasis of non-small cell lung carcinoma cells. RESULTS: Melanoma antigen family C2 was highly expressed in non-small cell lung carcinoma cells; melanoma antigen family C2 promoted the expression of epithelial-mesenchymal transition-related proteins as well as enhance the secretion of vascular endothelial growth factor and promote angiogenesis; melanoma antigen family C2 promoted proliferation, migration, and invasion and suppressed apoptosis of non-small cell lung carcinoma cells. It could also facilitate growth and metastasis of non-small cell lung carcinoma in vivo. CONCLUSION: Melanoma antigen family C2 was a critical factor of angiogenesis and epithelial-mesenchymal transition in non-small cell lung carcinoma.


Assuntos
Antígenos de Neoplasias/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Transição Epitelial-Mesenquimal/genética , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Animais , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , RNA Interferente Pequeno/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Artigo em Chinês | WPRIM | ID: wpr-701170

RESUMO

AIM:To observe whether modified epitopes from hepatocellular carcinoma antigen MAGEC 2 have HLA-A2-restricted antitumor ability.METHODS:HLA-A2 epitopes from MAGEC2 protein were predicted by NetCTL 1. 2,SYFPEITHI and IEDB.The change of binding anchor motifs by replacing anchor residues created the modified peptides from MAGEC2.The binding affinity of the peptides to HLA-A*0201 molecule was evaluated by T2 cells binding assay. ELISPOT assay and intracellular cytokine staining were used to investigate the ability of the peptides inducing specific re -stricted cytotoxic T-lymphocytes(CTLs)to release interferon-γ(IFN-γ).The ability of the peptides to induce T-cell re-sponse was investigated by cytotoxicity assay in vitro.RESULTS:The candidate peptides P248, P248-1Y,P356,P356-1Y,P356-2L and P356-1Y2L showed moderate affinity toward HLA-A2 molecule.T2 binding assay showed that P248-1Y and P356-1Y2L showed significantly higher affinity for HLA-A2 than the native peptides.ELISPOT assay and intracellular cytokine staining showed P248, P248-1Y, P356 and P356-1Y2L were able to induce specific CTLs to release IFN-γ. ELISPOT assay showed that significantly higher levels of IFN-γrelease were induced by P248-1Y and P356-1Y2L than the native peptides.The CTLs induced by P248,P248-1Y,P356 and P356-1Y2L lysed HepG2 cells,and P248-1Y and P356-1Y2L peptide-specific CTLs showed higher cytotoxicity against HepG 2 cells than the native peptide-specific CTLs(P<0.05).CONCLUSION: Compared with the native peptides, modified epitopes P248-1Y and P356-1Y2L have higher binding affinity with HLA-A2 and retain immunogenecity.In addition, the antitumor immunity effects of modified epitope P248-1Y and P356-1Y2L are stronger than the native peptides.The peptides P248-1Y and P356-1Y2L are excellent HLA-A2-restricted CTL epitopes from tumor antigen MAGEC 2, which could serve as new candidates towards antitumor peptide vaccines.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa