Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Cancer ; 15(1): 232-250, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38164271

RESUMO

Background: Insulin-like growth factor binding protein 5 (IGFBP5) is highly expressed in multiple human cancers, including glioma. Despite this, it remains unclear what role it plays in glioma. The aim of the present study was to analyze whether IGFBP5 could be used as a predictor of prognosis and immune infiltration in glioma. Methods: Glioma patients' clinical information was collected from the Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), Rembrandt, and Gravendeel databases. The diagnostic and prognostic roles of IGFBP5 were assessed by the Kaplan-Meier survival curves, diagnostic receiver operating characteristic (ROC) curves, nomogram model, Cox regression analysis and Enrichment analysis by R software. Moreover, the correlation between IGFBP5 expression and immune cell infiltration, and immune checkpoint genes was conducted. Immunohistochemistry staining, CCK8, colony formation, scratch and transwell assays and western blot were used to interrogate the expression and function of IGFBP5 in glioma. Results: IGFBP5 levels were obviously increased in glioma with higher malignancy and predicted poor outcomes by Univariate and multivariate Cox analysis. The biological function analysis revealed that IGFBP5 correlated closely with immune signatures. Moreover, IGFBP5 expression was associated with tumor infiltration of B cells, T cells, macrophages, and NK cells. IGFBP5 affected glioma cell proliferation, migration, and invasion probably involved in the epithelial-to-mesenchymal transition (EMT) and Hippo-YAP signaling pathway. Further study showed that IGFBP5 induced the expression of PD-L1 and CXCR4. Conclusions: IGFBP5 as an oncogene is a useful biomarker of prognosis and correlates with progression and immune infiltration in glioma.

2.
Med Sci Monit ; 29: e938660, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36973995

RESUMO

BACKGROUND Cervical cancer is one of the most common malignances among women globally. This study aimed to construct a novel immune-related signature to predict the prognosis and immune infiltration of cervical cancer. MATERIAL AND METHODS Transcriptomic profiles and corresponding clinical information of cervical cancer patients were obtained from The Cancer Genome Atlas (TCGA) database and GEO database. The hub immune-related genes were screened and selected using Cox regression analysis and LASSO regression analysis. A novel signature was established based on the expression levels and corresponding coefficients of the selected hub immune-related genes. Kaplan-Meier survival curve and ROC curve illustrated the prognostic value of this novel signature in cervical cancer. The predictive accuracy and stability of this novel signature were confirmed in the validation cohort, internal testing set and external testing set. Then, a nomogram was constructed to predict individual survival probability of cervical cancer patient. The association between the risk scores of novel signature and immune infiltration was investigated through single-sample gene set enrichment analysis (ssGSEA). RESULTS Ten hub immune-related genes (TFRC, SPP1, CAMP, CSF2, TUBB3, ZAP70, CHIT1, LEPR, DLL4, and DES) were selected to construct a novel signature. The risk score of this novel signature could be an independent prognostic factor in cervical cancer, which divided patients into high-risk and low-risk groups. The patients in high-risk groups showed significantly worse overall survival rates than those in low-risk groups in all training and validation cohorts (all P<0.05). A nomogram model was constructed based on the risk score of the novel signature and other clinical characteristics, which achieved the highest clinical net benefit across the entire range of reasonable threshold probabilities (concordance index=0.813). Furthermore, gene enrichment analysis revealed that the novel signature was closely related with immunology. The novel signature was negatively correlated with the infiltration of most immune cell types, especially T cell subsets (P<0.001). CONCLUSIONS The novel signature could comprehensively predict the prognosis and immune infiltration of cervical cancer. It may provide new insights for the precise treatment in cervical cancer.


Assuntos
Neoplasias do Colo do Útero , Humanos , Feminino , Neoplasias do Colo do Útero/genética , Prognóstico , Nomogramas , Fatores de Risco , Bases de Dados Factuais
3.
Cells ; 11(18)2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36139447

RESUMO

As a critical transformational process in the attributes of epithelial cells, epithelial-to-mesenchymal transition (EMT) is involved in tumor invasion, metastasis, and resistance to treatment, which contributes to the ultimate death of some patients with breast cancer. Glycogen synthase kinase-3-beta (GSK3ß) is thought to be an EMT suppressor that down-regulates the protein, snail, a zinc finger transcription inhibitor, and regulates E-cadherin expression and the Wnt signaling pathway. Our previous studies have shown that Notch3 also inhibits EMT in breast cancer. In mammary gland cells, GSK3ß physically bound and phosphorylated the intracellular domain of two Notch paralogs: N1ICD was positively regulated, but N2ICD was negatively regulated; however, the relationship between Notch3, GSK3ß, and EMT in breast cancer is still unclear and crosstalk between Notch3 and GSK3ß has not been widely investigated. In this study, we revealed that Notch3 was an essential antagonist of EMT in breast cancer cells by transcriptionally upregulating GSK3ß. In breast cancer, MCF-7 and MDA-MB-231 cell lines, the silencing of Notch3 reduced GSK3ß expression, which is sufficient to induce EMT. Conversely, ectopic Notch3 expression re-activated GSK3ß and E-cadherin. Mechanistically, Notch3 can bind to the GSK3ß promoter directly and activate GSK3ß transcription. In human breast cancer samples, Notch3 expression is positively associated with GSK3ß (r = 0.416, p = 0.001); moreover, high expressions of Notch3 and GSK3ß mRNA are correlated to better relapse-free survival in all breast cancer patients via analysis in "the Kaplan-Meier plotter" database. In summary, our preliminary results suggested that Notch3 might inhibit EMT by trans-activating GSK3ß in breast cancer cells. The suppression of Notch3 expression may contribute to EMT by transcriptionally downregulating GSK3ß in breast cancer.


Assuntos
Neoplasias da Mama , Transição Epitelial-Mesenquimal , Glicogênio Sintase Quinase 3 beta , Receptor Notch3 , Neoplasias da Mama/patologia , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Feminino , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Recidiva Local de Neoplasia , RNA Mensageiro , Receptor Notch3/genética , Via de Sinalização Wnt
4.
Metabolites ; 12(6)2022 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-35736417

RESUMO

Okra is a kind of flavonoid-rich food which was reported to have a variety of health functions. Flavonoids are the major polyphenolic compounds in okra and are thought to play a role in reducing the risk of disease. The aim of this study was to isolate and identify the flavonoids composition in okra pods and explore the activity of the main flavonoids components identified on inhibiting tumor cell proliferation in vitro. Six individual flavonoids were identified by HPLC-MS/MS: quercetin-3-gentiobioside (Q3G), quercetin-3-sambubioside (Q3S), rutin, quercetin-7-glucoside (Q7G), isoquercitrin (ISO) and quercetin-3-malonylglucoside (Q3M), which were all separated well within 30 min. The analytical method was validated by the recovery of spiked samples and so on. Moreover, four main flavonoids components, namely Q3G, Q3S, ISO and Q3M, exhibited significant (p < 0.05) inhibition of NCI-N87, A375, A549 cells proliferation (25−100 µmol/L) and of HFLS-RA (200−300 µmol/L) in different levels, according to MTT method, respectively. It is demonstrated that the flavonoids components of okra exhibited a noteworthy development prospect as a possible nutraceutical dietary supplement.

5.
Oncotarget ; 13: 583-584, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35391718

RESUMO

[This corrects the article DOI: 10.18632/oncotarget.2741.].

6.
Eur J Cell Biol ; 101(2): 151199, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35066432

RESUMO

Breast cancer (BC) and Alzheimer's disease (AD) have pronounced female-to-male disparities and both are the major causes of death in elderly women. Intriguingly, there is an inverse incidence between BC and AD. In our previous study, we found that the expression of ARSD, a female-biased gene on chromosome Xp22.3 that encodes arylsulfatase D, is significantly downregulated in triple-negative breast cancer (TNBC) cells and tissue samples, and that ectopic ARSD overexpression could inhibit proliferation and migration of BC cells. However, the exact mechanism remains unclear. In this study, ARSD-overexpressing MDA-MB-231 cell strains were established. RNA-Seq and qRT-PCR validation were performed followed by GO and KEGG analyses. Transcriptome sequencing unveiled that Alzheimer's/Parkinson's/prion diseases were enriched in ARSD overexpressing BC cells. Besides, the top enriched pathways included lipoprotein/cholesterol metabolism, molecular chaperone and misfolding protein binding, mitochondrial respiration, dysfunction of lysosomes, etc. In which, a battery of genes, e.g., SERF1A, APOE, CD36 etc., were upregulated, while a series of genes, e.g., NDUFA11, NDUFS3, NDUFV1, etc. were downregulated, which were closely related to amyloidosis. The amyloidosis of BC cells and nerval cells caused by ARSD overexpression was verified with western blotting, immunohistochemical and Congo red staining. Collectively, downregulated ARSD may be closely associated with BC, and upregulated ARSD may cause amyloidosis of BC cells. Our findings suggest that ARSD deserves to be considered a new promising target for treating TNBC or for AD.


Assuntos
Amiloidose , Neoplasias da Mama , Carcinoma , Neoplasias de Mama Triplo Negativas , Idoso , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células , Células Epiteliais/patologia , Feminino , Humanos , Masculino , Proteínas do Tecido Nervoso , Fatores de Transcrição , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
7.
Cell Death Dis ; 12(11): 1042, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34725332

RESUMO

Advanced breast cancer (BC), especially basal like triple-negative BC (TNBC), is a highly malignant tumor without viable treatment option, highlighting the urgent need to seek novel therapeutic targets. Arylsulfatase D (ARSD), localized at Xp22.3, is a female-biased gene due to its escaping from X chromosome inactivation (XCI). Unfortunately, no systematic investigation of ARSD on BC has been reported. In this study, we observed that ARSD expression was positively related to ERα status either in BC cells or tissue specimens, which were associated with good prognosis. Furthermore, we found a set of hormone-responsive lineage-specific transcription factors, FOXA1, GATA3, ERα, directly drove high expression of ARSD through chromatin looping in luminal subtype BC cells. Opposingly, ARSD still subjected to XCI in TNBC cells mediated by Xist, CpG islands methylation, and inhibitory histone modification. Unexpectedly, we also found that ectopic ARSD overexpression could inhibit proliferation and migration of TNBC cells by activating Hippo/YAP pathway, indicating that ARSD may be a molecule brake on ERα signaling pathway, which restricted ERα to be an uncontrolled active status. Combined with other peoples' researches that Hippo signaling maintained ER expression and ER + BC growth, we believed that there should exist a regulative feedback loop formation among ERα, ARSD, and Hippo/YAP pathway. Collectively, our findings will help filling the knowledge gap about the influence of ARSD on BC and providing evidence that ARSD may serve as a potential marker to predict prognosis and as a therapeutic target.


Assuntos
Arilsulfatases/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Movimento Celular/genética , Receptor alfa de Estrogênio/metabolismo , Via de Sinalização Hippo , Proteínas de Sinalização YAP , Arilsulfatases/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/genética , Cromatina/metabolismo , Cromossomos Humanos X/genética , Metilação de DNA/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Via de Sinalização Hippo/genética , Histonas/metabolismo , Humanos , Pessoa de Meia-Idade , Modelos Biológicos , Fenótipo , Processamento de Proteína Pós-Traducional , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Inativação do Cromossomo X , Proteínas de Sinalização YAP/genética
8.
Cell Death Dis ; 12(8): 779, 2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-34365463

RESUMO

The PD-L1 overexpression is an important event of immune escape and metastasis in triple-negative breast cancer (TNBC), but the molecular mechanism remains to be determined. Interferon gamma (IFNγ) represents a major driving force behind PD-L1 expression in tumor microenvironment, and histone deacetylase 2 (HDAC2) is required for IFN signaling. Here, we investigated the regulation of HDAC2 on the IFNγ-induced PD-L1 expression in TNBC cells. We found the HDAC2 and PD-L1 expression in TNBC was significantly higher than that in non-TNBC, and HDAC2 was positively correlated with PD-L1 expression. HDAC2 promoted PD-L1 induction by upregulating the phosphorylation of JAK1, JAK2, and STAT1, as well as the translocation of STAT1 to the nucleus and the recruitment of STAT1 to the PD-L1 promoter. Meanwhile, HDAC2 was recruited to the PD-L1 promoter by STAT1, and HDAC2 knockout compromised IFNγ-induced upregulation of H3K27, H3K9 acetylation, and the BRD4 recruitment in PD-L1 promoter. In addition, significant inhibition of proliferation, colony formation, migration, and cell cycle of TNBC cells were observed following knockout of HDAC2 in vitro. Furthermore, HDAC2 knockout reduced IFNγ-induced PD-L1 expression, lymphocyte infiltration, and retarded tumor growth and metastasis in the breast cancer mouse models. This study may provide evidence that HDAC2 promotes IFNγ-induced PD-L1 expression, suggesting a way for enhanced antitumor immunity when targeting the HDAC2 in TNBC.


Assuntos
Antígeno B7-H1/genética , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Histona Desacetilase 2/deficiência , Evasão da Resposta Imune , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Acetilação/efeitos dos fármacos , Animais , Antígeno B7-H1/metabolismo , Sequência de Bases , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Histona Desacetilase 2/metabolismo , Histonas/metabolismo , Humanos , Evasão da Resposta Imune/genética , Interferon gama/farmacologia , Janus Quinase 2/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Regiões Promotoras Genéticas/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
9.
Front Cell Dev Biol ; 8: 594372, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33244467

RESUMO

Our previous study found that Notch3 knockout mice exhibit defects in mammary gland development. To elucidate the underlying mechanism, tissue samples were subjected to RNA-seq, GO, and KEGG enrichment analyses and qRT-PCR validation. Of enriched pathways, chemokine signaling pathway and cytokine-cytokine receptor interaction were noticed in both Notch3wt/wt/Notch3wt/- and Notch3wt/wt/Notch3-/- mice, in which the expression of chemokine ligand 2 (CCL2) was sharply reduced in Notch3wt/- and Notch3-/- mammary gland tissues. The Mouse ENCODE transcriptome data reveal that the mammary gland fat pad exhibits a high CCL2, CCR2, and CCR4 expression, indicating that these molecules play important roles during mammary gland development. Specifically, defective mammary glands in Notch3 knockout mice could be partially rescued by CCL2 overexpression lentivirus through intraductal injection. An in vitro study showed that CCL2 overexpression promoted the proliferation, migration, and cancerous acinar formation of 4T1 cells, which could rescue the defective migration of 4T1 cells caused by Notch3 knockdown. We also found that Notch3 transcriptionally regulated the expression of CCL2 in a classical pattern. Our findings illustrated that Notch3-regulating CCL2/CCR4 axis should be an important signaling pathway for mammary gland development and should be a candidate target for breast cancer therapy.

10.
Exp Cell Res ; 386(2): 111743, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31770532

RESUMO

Notch3 can act as a tumor suppressor in the breast cancer epithelial cells. Unfortunately, Notch3 expression is decreased or lost, especially in triple-negative breast cancer (TNBC) cells, and the reasons remain unclear. Here, we found Notch3 was upregulated in MDA-MB-231 cells with 5-Aza treatment. Two CpG islands were observed in notch3 promoter. Interestingly, bisulfite sequencing exhibited that large amounts of unconverted cytosines were not only followed by guanine, but also adenine, cytosine and thymine, which implied that there simultaneously existed CpG and non-CpG methylation in notch3 promoter. To better analyze the methylation frequency of non-CpG locus, we designed CpG/non-CpG methylation analysis software. The results showed that the methylation frequency of notch3 gene in different breast cancer cell lines was in order T47D, MCF-7, SKBR3, BT-549 and MDA-MB-231. Furthermore, we identified that DNMT3b, DNMT1, DNMT3L, Mecp2 and EZH2 were important regulators of non-CpG locus of notch3 gene. Immunohistochemistry staining revealed a negative correlation between EZH2 and Notch3 from 22 luminal and 26 TNBC cases. In vitro methylation combined luciferase activity assays showed that non-CpG methylation was still crucial cause leading to notch3 transcriptional repression in TNBC. Our findings provide possible explanation for the downregulation or loss of Notch3 expression in TNBC.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas , Receptor Notch3/genética , Antimetabólitos Antineoplásicos/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Decitabina/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Genes Reporter , Humanos , Luciferases/genética , Luciferases/metabolismo , Células MCF-7 , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Receptor Notch3/deficiência , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , DNA Metiltransferase 3B
11.
J Cell Mol Med ; 24(1): 1116-1127, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31755192

RESUMO

Adipocytes constitute a major component of the tumour microenvironment. Numerous studies have shown that adipocytes promote aggressiveness and invasion by stimulating cancer cells proliferation and modulating their metabolism. Herein, we reported that Notch3 promotes mouse 3T3-L1 pre-adipocytes differentiation by performing the integrative transcriptome and TMT-based proteomic analyses. The results revealed that aminoacyl-tRNA_biosynthesis pathway was significantly influenced with Nocth3 change during 3T3-L1 pre-adipocytes differentiation, and the expression of LARS in this pathway was positively correlated with Notch3. Published studies have shown that LARS is a sensor of leucine that regulates the mTOR pathway activity, and the latter involves in adipogenesis. We therefore supposed that Notch3 might promote 3T3-L1 pre-adipocytes differentiation by up-regulating LARS expression and activating mTOR pathway. CHIP and luciferase activity assay uncovered that Notch3 could transcriptionally regulate the expression of LARS gene. Oil Red staining identified a positive correlation between Notch3 expression and adipocytic differentiation. The activation of mTOR pathway caused by Notch3 overexpression could be attenuated by knocking down LARS expression. Altogether, our study revealed that Notch3 promotes adipocytic differentiation of 3T3-L1 pre-adipocytes cells by up-regulating LARS expression and activating the mTOR pathway, which might be an emerging target for obesity treatment.


Assuntos
Adipócitos/citologia , Adipogenia , Diferenciação Celular , Regulação da Expressão Gênica , Leucina-tRNA Ligase/metabolismo , Receptor Notch3/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Biomarcadores/análise , Leucina-tRNA Ligase/genética , Camundongos , Proteoma/análise , Receptor Notch3/genética , Serina-Treonina Quinases TOR/genética , Transcriptoma
12.
BioData Min ; 12: 17, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31462928

RESUMO

BACKGROUND: The clinical outcomes of patients with resected T1-3N0-2M0 non-small cell lung cancer (NSCLC) with the same tumor-node-metastasis (TNM) stage are diverse. Although other prognostic factors and prognostic prediction tools have been reported in many published studies, a convenient, accurate and specific prognostic prediction software for clinicians has not been developed. The purpose of our research was to develop this type of software that can analyze subdivided T and N staging and additional factors to predict prognostic risk and the corresponding mean and median survival time and 1-5-year survival rates of patients with resected T1-3N0-2M0 NSCLC. RESULTS: Using a Cox proportional hazard regression model, we determined the independent prognostic factors and obtained a prognostic index (PI) eq. PI = ∑ßixi.=0.379X1-0.403X2-0.267X51-0.167X61-0.298X62 + 0.460X71 + 0.617X72-0.344X81-0.105X91-0.243X92 + 0.305X101 + 0.508X102 + 0.754X103 + 0.143X111 + 0.170X112 + 0.434X113-0.327X122-0.247X123 + 0.517X133 + 0.340X134 + 0.457X143 + 0.419X144 + 0.407X145. Using the PI equation, we determined the PI value of every patient. According to the quantile of the PI value, patients were divided into three risk groups: low-, intermediate-, and high-risk groups with significantly different survival rates. Meanwhile, we obtained the mean and median survival times and 1-5-year survival rates of the three groups. We developed the RNSCLC-PRSP software which is freely available on the web at http://www.rnsclcpps.com with all major browsers supported to determine the prognostic risk and associated survival of patients with resected T1-3N0-2 M0 non-small cell lung cancer. CONCLUSIONS: After prognostic factor analysis, prognostic risk grouping and corresponding survival assessment, we developed a novel software program. It is practical and convenient for clinicians to evaluate the prognostic risk and corresponding survival of patients with resected T1-3N0-2M0 NSCLC. Additionally, it has guiding significance for clinicians to make decisions about complementary treatment for patients.

13.
Cell Adh Migr ; 13(1): 203-218, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31096822

RESUMO

EMT allows a polarized epithelium to lose epithelial integrity and acquire mesenchymal characteristics. Previously, we found that overexpression of the intracellular domain of Notch3 (N3ICD) can inhibit EMT in breast cancer cells. In this study, we aimed to elucidate the influence of N3ICD or N3ICD combined with the transmembrane domain (TD+N3ICD) on the expression and distribution of TJs/AJs and polar molecules. We found that although N3ICD can upregulate the expression levels of the above-mentioned molecules, TD+N3ICD can inhibit EMT more effectively than N3ICD alone. TD+N3ICD overexpression upregulated the expression of endogenous full-length Notch3 and contributed to correcting the position of TJs/AJs molecules and better acinar structures formation. Co-immunoprecipitation results showed that the upregulated endogenous full-length Notch3 could physically interact with E-ca in MDA-MB-231/pCMV-(TD+N3ICD) cells. Collectively, our data indicate that overexpression of TD+N3ICD can effectively inhibit EMT, resulting in better positioning of TJs/AJs molecules and cell-cell adhesion in breast cancer cells. Abbreviations: EMT: Epithelial-mesenchymal transition; TJs: Tight junctions; AJs: Adherens junctions; aPKC: Atypical protein kinase C; Crb: Crumbs; Lgl: Lethal (2) giant larvae; LLGL2: lethal giant larvae homolog 2; PAR: Partitioning defective; PATJ: Pals1-associated TJ protein.


Assuntos
Junções Aderentes/patologia , Neoplasias da Mama/patologia , Membrana Celular , Transição Epitelial-Mesenquimal , Receptor Notch3/metabolismo , Junções Íntimas/patologia , Junções Aderentes/metabolismo , Neoplasias da Mama/metabolismo , Adesão Celular , Polaridade Celular , Feminino , Humanos , Domínios Proteicos , Receptor Notch3/genética , Junções Íntimas/metabolismo , Células Tumorais Cultivadas
14.
Oncogenesis ; 8(4): 26, 2019 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-30940798

RESUMO

HIF-1α has a broad impact on tumors, including enhanced utilization of glucose, tumor cell stemness, migration, metastasis and so on. In pilot study, we found that the expression of HIF-1α significantly increased in breast cancer cell lines and tissue samples with higher malignant behaviors and decreased in luminal subtype breast cancer cells and tissue samples. We analyzed and found there is one large CpG island in HIF-1α promoter around transcription start site, and the hypermethylation occurred at these CpGs and their surrounding non-CpGs sites. Epigenetic events driving tumorigenesis has been characterized. However, knowledge is lacking on the non-CpGs methylation of HIF-1α promoter in breast cancer cells. We validated that non-CpGs methylation can directly regulate HIF-1α expression by luciferase activity assay. We also found DNMT3a and Mecp2 play vital role in methylation at non-CpGs and CpGs sites. In addition, we noticed that H3K9ac modification could promote the transcription of HIF-1α in MDA-MB-231 cells by binding to the region contained hypomethylated non-CpG and CpG sites. Taken together, the hypomethylation status at non-CpG and CpG loci in HIF-1α promoter and H3K9ac modification together contribute to maintain higher HIF-1αactivity in invasive breast cancer cells when compared with the non-invasive breast cancer cells, which may establish a tissue-specific epigenetic modification pattern and point to the new directions for future understanding breast cancer therapy.

15.
Cell Death Dis ; 9(12): 1171, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30518916

RESUMO

Recent studies indicate that the long noncoding RNA ATB (lncATB) can induce the epithelial-mesenchymal transition (EMT) in cancer cells, but the specific cellular targets of lncATB require further investigation. In the present study, the upregulation of lncATB in breast cancer cells was validated in a TGF-ß-induced EMT model. Gain- and loss-of-function studies demonstrated that lncATB enhanced cell migration, invasion and clonogenicity in vitro and in vivo. LncATB promoted the EMT by acting as a sponge for the miR-200 family and restoring Twist1 expression. Subsequently, the clinical significance of lncATB was investigated in a cohort of breast cancer patients (N = 131). Higher lncATB expression was correlated with increased nodal metastasis (P = 0.036) and advanced clinical stage (P = 0.011) as well as shorter disease-free survival (P = 0.043) and overall survival (P = 0.046). These findings define Twist1 as a major target of lncATB in the induction of the EMT and highlight lncATB as a biomarker in breast cancer patients.


Assuntos
Neoplasias da Mama/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Proteínas Nucleares/genética , RNA Longo não Codificante/genética , Proteína 1 Relacionada a Twist/genética , Adulto , Animais , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Estudos de Coortes , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Metástase Linfática , Células MCF-7 , Camundongos , Camundongos Nus , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Proteínas Nucleares/metabolismo , Prognóstico , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Fator de Crescimento Transformador beta/farmacologia , Carga Tumoral , Proteína 1 Relacionada a Twist/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Cell Signal ; 51: 243-256, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30121333

RESUMO

It is well known that epithelial-mesenchymal transition (EMT) can confer cancer cells with invasive and migratory capabilities associated with distant metastasis. As a key upstream factor in the Hippo pathway, Kibra (wwc1 gene) has been shown to suppress EMT in breast cancer cells, and we have found that its expression is reduced or lost completely in both human breast cancer cell lines and clinical tissue samples, particularly in triple negative breast cancer (TNBC). Unfortunately, the molecular mechanisms underlying this progression-associated event remain to be elucidated. Epigenetic gene silencing is one of the most common causes of suppressed expression of tumor suppressor genes. Furthermore, recent studies have demonstrated that EZH2 can recruit DNA methyltransferases, resulting in DNA methylation and subsequent gene silencing in certain circumstances. Thus, we hypothesized that there may exist a link between EZH2 and DNA methylation in association with wwc1 silencing in breast cancer. To test this hypothesis, we performed bisulfite sequencing, shRNA, co-IP, ChIP, MeDIP and ChIP-qPCR. As expected, RG108 or 5-Aza treatment improved the wwc1 gene transcription and Kibra protein expression. Both bisulfite sequencing and MeDIP demonstrated higher CpG methylation of the wwc1 promoter the TNBC cells (MDA-MB-231) than in luminal breast cancer cells (MCF7). It is noteworthy that ChIP and co-IP assays showed that EZH2, H3K27me3 and DNMT1 are enriched at the wwc1 promoter, and there exist physiologically relevant protein-protein interactions between them. We also found that EZH2 knockdown leads to a partial increase in Kibra expression and a considerable reduction in H3K27 and DNMT1 trimethylation. Moreover, ChIP-qPCR revealed more DNA fragments containing the wwc1 promoter in MDA-MB-231 than in MCF7 cells after immunoprecipitation with EZH2, DNMT1 and H3K27me3 antibodies. Collectively, our results reveal crosstalk between H3K27me3 inhibition catalyzed by EZH2 and CpG island methylation mediated by DNMT1 within the wwc1 promoter, which synergistically silence wwc1 gene expression in TNBC. Based on these results, we conclude that EZH2 shows promise as a potential anti-tumor target.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Histona Desmetilases com o Domínio Jumonji/fisiologia , Fosfoproteínas/genética , Neoplasias de Mama Triplo Negativas/genética , Movimento Celular , Ilhas de CpG , Metilação de DNA , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Feminino , Via de Sinalização Hippo , Humanos , Células MCF-7 , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
17.
Oncogenesis ; 7(8): 59, 2018 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-30100605

RESUMO

Notch3 and GATA binding protein 3 (GATA-3) have been, individually, shown to maintain luminal phenotype and inhibit epithelial-mesenchymal transition (EMT) in breast cancers. In the present study, we report that Notch3 expression positively correlates with that of GATA-3, and both are associated with estrogen receptor-α (ERα) expression in breast cancer cells. We demonstrate in vitro and in vivo that Notch3 suppressed EMT and breast cancer metastasis by activating GATA-3 transcription. Furthermore, Notch3 knockdown downregulated GATA-3 and promoted EMT; while overexpression of Notch3 intracellular domain upregulated GATA-3 and inhibited EMT, leading to a suppression of metastasis in vivo. Moreover, inhibition or overexpression of GATA-3 partially reversed EMT or mesenchymal-epithelial transition induced by Notch3 alterations. In breast cancer patients, high GATA-3 expression is associated with higher Notch3 expression and lower lymph node metastasis, especially for hormone receptor (HR) positive cancers. Herein, we demonstrate a novel mechanism whereby Notch3 inhibit EMT by transcriptionally upregulating GATA-3 expression, at least in part, leading to the suppression of cancer metastasis in breast cancers. Our findings expand our current knowledge on Notch3 and GATA-3's roles in breast cancer metastasis.

18.
Theranostics ; 7(16): 4041-4056, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29109797

RESUMO

The luminal A phenotype is the most common breast cancer subtype and is characterized by estrogen receptor α expression (ERα). Identification of the key regulator that governs the luminal phenotype of breast cancer will clarify the pathogenic mechanism and provide novel therapeutic strategies for this subtype of cancer. ERα signaling pathway sustains the epithelial phenotype and inhibits the epithelial-mesenchymal transition (EMT) of breast cancer. In this study, we demonstrate that Notch3 positively associates with ERα in both breast cancer cell lines and human breast cancer tissues. We found that overexpression of Notch3 intra-cellular domain, a Notch3 active form (N3ICD), in ERα negative breast cancer cells re-activated ERα, while knock-down of Notch3 reduced ERα transcript and proteins, with alteration of down-stream genes, suggesting its ability to regulate ERα. Mechanistically, our results show that Notch3 specifically binds to the CSL binding element of the ERα promoter and activates ERα expression. Moreover, Notch3 suppressed EMT, while suppression of Notch3 promoted EMT in cellular assay. Overexpressing N3ICD in triple-negative breast cancer suppressed tumorigenesis and metastasis in vivo. Conversely, depletion of Notch3 in luminal breast cancer promoted metastasis in vivo. Furthermore, Notch3 transcripts were significantly associated with prolonged relapse-free survival in breast cancer, in particular in ERα positive breast cancer patients. Our observations demonstrate that Notch3 governs the luminal phenotype via trans-activating ERα expression in breast cancer. These findings delineate the role of a Notch3/ERα axis in maintaining the luminal phenotype and inhibiting tumorigenesis and metastasis in breast cancer, providing a novel strategy to re-sensitize ERα negative or low-expressing breast cancers to hormone therapy.


Assuntos
Neoplasias da Mama/metabolismo , Receptor Notch3/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/genética , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor Notch3/genética , Receptores de Estrogênio/genética
19.
Methods Mol Biol ; 1652: 259-274, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28791647

RESUMO

Breast cancer, the most common malignancy among women worldwide, is a heterogeneous disease, and it therefore has remarkably different biological characteristics and clinical behavior. Breast cancer has been divided into several different molecular subtypes based on the status of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor 2 (HER2, also named as ErbB2) status. Her2 is a member of EGFR family of transmembrane tyrosine kinase-type receptors, and is involved in the activation of its downstream signaling cascades, which could promote cell proliferation, metastasis, and angiogenesis in tumors. In addition, Twist, a transcriptional factor has been shown to associate with ErbB2 signaling to increase the proliferation and the number of cells, and to induce epithelial-mesenchymal transition. Deregulated cell proliferation can result in hyperplasia and even malignancies. Actually, the proliferative or survival ability of cells can be measured by a variety of methods. Clonogenic assay and CCK8 assay can serve as useful tools to test whether the clonogenic survival ability of tumor cells can be enhanced or reduced upon stimulation of appropriate mitogenic signals or a given cancer therapy respectively. A colony is defined as a cluster of at least 50 cells that can often only be determined microscopically. Moreover, migration and invasion assay, in some degree, represents the potential for EMT promotion. Here, we introduce colony formation assay; CCK8 proliferation assay; soft agar; and migration and invasion assay using overexpression of ErbB2 and EGFR receptors as an example.


Assuntos
Bioensaio , Transição Epitelial-Mesenquimal/genética , Expressão Gênica , Animais , Bioensaio/métodos , Células COS , Movimento Celular/genética , Proliferação de Células , Chlorocebus aethiops , Ensaio de Unidades Formadoras de Colônias , Técnicas In Vitro , Fatores de Transcrição Twist
20.
Cancer Lett ; 386: 65-76, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27838413

RESUMO

Tamoxifen resistance presents a prominent clinical challenge in endocrine therapy for hormone sensitive breast cancer. However, the underlying mechanisms that contribute to tamoxifen resistance are not fully understood. In this study, we established a tamoxifen resistant MCF-7 cell line (MCF-7-Tam-R) by continuously incubating MCF-7 cells with 4-OH-tamoxifen. We found that melanoma cell adhesion molecule (MCAM/CD146), a unique epithelial-to-mesenchymal transition (EMT) inducer, was significantly up-regulated at both mRNA and protein levels in MCF-7-Tam-R cells compared to parental MCF-7 cells. Mechanistic research demonstrated that MCAM promotes tamoxifen resistance by transcriptionally suppressing ERα expression and activating the AKT pathway, followed by induction of EMT. Elevated MCAM expression was inversely correlated with recurrence-free and distant metastasis-free survival in a cohort of 4142 patients with breast cancer derived from a public database, particularly in the subgroup only treated with tamoxifen. These results demonstrate a novel function of MCAM in conferring tamoxifen resistance in breast cancer. Targeting MCAM might be a promising therapeutic strategy to overcome tamoxifen resistance in breast cancer patients.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tamoxifeno/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Antígeno CD146/genética , Antígeno CD146/metabolismo , Movimento Celular/efeitos dos fármacos , Bases de Dados Factuais , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Regulação para Baixo , Ativação Enzimática , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Células MCF-7 , Metástase Neoplásica , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA