Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
Sci Rep ; 14(1): 12477, 2024 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816533

RESUMO

Dysregulated Wnt/ß-catenin signaling is a common feature of colorectal cancer (CRC). The T-cell factor/lymphoid enhancer factor (TCF/LEF; hereafter, TCF) family of transcription factors are critical regulators of Wnt/ß-catenin target gene expression. Of the four TCF family members, TCF7L1 predominantly functions as a transcriptional repressor. Although TCF7L1 has been ascribed an oncogenic role in CRC, only a few target genes whose expression it regulates have been characterized in this cancer. Through transcriptome analyses of TCF7L1 regulated genes, we noted enrichment for those associated with cellular migration. By silencing and overexpressing TCF7L1 in CRC cell lines, we demonstrated that TCF7L1 promoted migration, invasion, and adhesion. We localized TCF7L1 binding across the CRC genome and overlapped enriched regions with transcriptome data to identify candidate target genes. The growth arrest-specific 1 (GAS1) gene was among these and we demonstrated that GAS1 is a critical mediator of TCF7L1-dependent CRC cell migratory phenotypes. Together, these findings uncover a novel role for TCF7L1 in repressing GAS1 expression to enhance migration and invasion of CRC cells.


Assuntos
Movimento Celular , Neoplasias Colorretais , Regulação Neoplásica da Expressão Gênica , Proteína 1 Semelhante ao Fator 7 de Transcrição , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Movimento Celular/genética , Linhagem Celular Tumoral , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Invasividade Neoplásica , Adesão Celular/genética , Via de Sinalização Wnt
2.
Oncol Res ; 32(3): 463-476, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38370338

RESUMO

An important factor in the emergence and progression of osteosarcoma (OS) is the dysregulated expression of microRNAs (miRNAs). Transcription factor 7-like 1 (TCF7L1), a member of the T cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor family, interacts with the Wnt signaling pathway regulator ß-catenin and acts as a DNA-specific binding protein. This study sought to elucidate the impact of the interaction between miR-329-3p and TCF7L1 on the growth and apoptosis of OS and analyze the regulatory expression relationship between miRNA and mRNA in osteosarcoma cells using a variety of approaches. MiR329-3p was significantly downregulated, while TCF7L1 was considerably up-regulated in all examined OS cell lines. Additionally, a clinical comparison study was performed using the TCGA database. Subsequently, the regulatory relationship between miR-329-3p and TCF7L1 on the proliferation and apoptosis of OS cells was verified through in vitro and in vivo experiments. When miR-329-3p was transfected into the OS cell line, the expression of TCF7L1 decreased, the proliferation of OS cells was inhibited, the cytoskeleton disintegrated, and the nucleus condensed to form apoptotic bodies. The expression of proteins that indicate apoptosis increased simultaneously. The cell cycle was arrested in the G0/G1 phase, and the G1/S transition was blocked. The introduction of miR-329-3p also inhibited downstream Cyclin D1 of the Wnt pathway. Xenograft experiments indicated that the overexpression of miR-329-3p significantly inhibited the growth of OS xenografts in nude mice, and the expression of TCF7L1 and c-Myc in tumor tissues decreased. MiR-329-3p was significantly reduced in OS cells and played a suppressive role in tumorigenesis and proliferation by targeting TCF7L1 both in vitro and in vivo. Osteosarcoma cell cycle arrest and pathway inhibition were observed upon the regulation of TCF7L1 by miR-329-3p. Summarizing these results, it can be inferred that miR-329-3p exerts anticancer effects in osteosarcoma by inhibiting TCF7L1.


Assuntos
Neoplasias Ósseas , MicroRNAs , Osteossarcoma , Proteína 1 Semelhante ao Fator 7 de Transcrição , Via de Sinalização Wnt , Animais , Humanos , Camundongos , beta Catenina/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Osteossarcoma/patologia , Via de Sinalização Wnt/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética
3.
Genes (Basel) ; 14(2)2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36833408

RESUMO

Mutations in components of the Wnt/ß-catenin signaling pathway drive colorectal cancer (CRC), in part, by deregulating expression of genes controlled by the T-cell factor (TCF) family of transcription factors. TCFs contain a conserved DNA binding domain that mediates association with TCF binding elements (TBEs) within Wnt-responsive DNA elements (WREs). Intestinal stem cell marker, leucine-rich-repeat containing G-protein-coupled receptor 5 (LGR5), is a Wnt target gene that has been implicated in CRC stem cell plasticity. However, the WREs at the LGR5 gene locus and how TCF factors directly regulate LGR5 gene expression in CRC have not been fully defined. Here, we report that TCF family member, TCF7L1, plays a significant role in regulating LGR5 expression in CRC cells. We demonstrate that TCF7L1 binds to a novel promoter-proximal WRE through association with a consensus TBE at the LGR5 locus to repress LGR5 expression. Using CRISPR activation and interference (CRISPRa/i) technologies to direct epigenetic modulation, we demonstrate that this WRE is a critical regulator of LGR5 expression and spheroid formation capacity of CRC cells. Furthermore, we found that restoring LGR5 expression rescues the TCF7L1-mediated reduction in spheroid formation efficiency. These results demonstrate a role for TCF7L1 in repressing LGR5 gene expression to govern the spheroid formation potential of CRC cells.


Assuntos
Neoplasias Colorretais , Receptores Acoplados a Proteínas G , Proteína 1 Semelhante ao Fator 7 de Transcrição , Humanos , beta Catenina/genética , Neoplasias Colorretais/genética , Células-Tronco Neoplásicas/metabolismo , Receptores Acoplados a Proteínas G/genética , Fatores de Transcrição/metabolismo
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(1): 66-73, 2022 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-35078577

RESUMO

Objective To investigate the expression of T cell factor 3 (TCF3) in hepatocellular carcinoma (HCC), its correlation with the prognosis of HCC patients, and its effect on the invasion, migration, and metastasis of HCC cells. Methods The expression of TCF3 mRNA in HCC tissues was detected with tumor public databases and the expression of TCF3 protein in HCC specimens was detected by immunohistochemical staining. Correlation between TCF3 expression and HCC patients' prognosis was analyzed. Western blot analysis was used to detect the expression of TCF3 in different human HCC cell lines, and lentivirus infection was conducted to construct TCF3-upregulated and TCF3-downregulated HCC cell lines. The effect of TCF3 on the invasion and migration of HCC cells was assessed by in vitro TranswellTM assay, and in vivo intrahepatic tumor implantation models were established to evaluate the effect of TCF3 on the metastatic capacity of HCC cells. Results The expression of TCF3 mRNA was significantly higher in HCC tissues than that in normal liver tissues, and high expression of TCF3 mRNA was closely correlated with decreased overall survival rates of HCC patients. In 120 cases of HCC tissues, the protein level of TCF3 was significantly higher than that in adjacent nontumor tissues, and patients with positive TCF3 expression had a markedly decreased overall survival rate and a higher recurrence rate compared with patients with negative TCF3 expression. In vitro TranswellTM assay indicated that TCF3 upregulation promoted the invasion and migration of PLC/PRF/5 cells, whereas knockdown of TCF3 inhibited the invasion and migration abilities of HCCLM3 cells. Intrahepatic tumor implantation models showed that TCF3 upregulation promoted the metastasis of PLC/PRF/5 cells, while TCF3 knockdown weakened the metastatic capacity of HCCLM3 cells. Conclusion TCF3 expression is significantly upregulated in human HCC tissues, and high TCF3 expression predicts a poor prognosis of HCC patients. TCF3 markedly promotes the invasion, migration, and metastasis of HCC cells.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Invasividade Neoplásica , Metástase Neoplásica
5.
Cell Rep ; 36(11): 109705, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525377

RESUMO

Mouse embryonic stem cells (mESCs) can be maintained in vitro in defined N2B27 medium supplemented with two chemical inhibitors for GSK3 and MEK (2i) and the cytokine leukemia inhibitory factor (LIF), which act synergistically to promote self-renewal and pluripotency. Here, we find that genetic deletion of the four genes encoding the TCF/LEF transcription factors confers mESCs with the ability to self-renew in N2B27 medium alone. TCF/LEF quadruple knockout (qKO) mESCs display dysregulation of several genes, including Aire, Dnmt3l, and IcosL, located adjacent to each other within a topologically associated domain (TAD). Aire, Dnmt3l, and IcosL appear to be regulated by TCF/LEF in a ß-catenin independent manner. Moreover, downregulation of Aire and Dnmt3l in wild-type mESCs mimics the loss of TCF/LEF and increases mESC survival in the absence of 2iL. Hence, this study identifies TCF/LEF effectors that mediate exit from the pluripotent state.


Assuntos
Autorrenovação Celular , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1 de Ligação ao Facilitador Linfoide/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Animais , Benzamidas/farmacologia , Autorrenovação Celular/efeitos dos fármacos , Meios de Cultura/química , Meios de Cultura/farmacologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Edição de Genes , Fator 1-alfa Nuclear de Hepatócito/deficiência , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Ligante Coestimulador de Linfócitos T Induzíveis/antagonistas & inibidores , Ligante Coestimulador de Linfócitos T Induzíveis/genética , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/deficiência , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/deficiência , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/deficiência , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , beta Catenina/deficiência , beta Catenina/genética , Proteína AIRE
6.
Biomed Res Int ; 2021: 6670456, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33824876

RESUMO

BACKGROUND: Cervical cancer (CC) is the second most common tumor in women worldwide. Studies have been accepted that genetic variations play an important role in the development of CC. The aim of this study was to evaluate the impact of TCF7L1 variants on CC risk. METHODS: 508 patients of cervical cancer and 497 healthy subjects were recruited to determine the impact of TCF7L1 polymorphisms on CC susceptibility. The associations were investigated by computing odds ratios (ORs) and 95% confidence intervals. The effect of SNP-SNP interactions on CC risk was explored by multifactor dimensionality reduction analysis. RESULTS: Our study showed that rs11904127 (OR 0.79, p = 0.010) and rs62162674 (OR 0.82, p = 0.044) of TCF7L1 significantly decreased cervical cancer risk. Stratified analysis indicated that rs11904127 and rs62162674 present decreased susceptibility to CC in age > 51 years (OR 0.74, p = 0.019; OR 0.72, p = 0.014, respectively). Haplotype analyses revealed that Grs2366264Trs11689667Crs62162674 has a lower risk to cervical cancer (OR = 0.43, p = 0.018). Besides, there is strong interaction of rs11904127 and rs2366264. CONCLUSION: Rs11904127 and rs62162674 in TCF7L1 are related to cervical cancer. We suggest that these variants can be used as prognostic markers for judging the susceptibility to cervical cancer.


Assuntos
Povo Asiático/genética , Predisposição Genética para Doença , Proteínas de Neoplasias/genética , Polimorfismo de Nucleotídeo Único , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Neoplasias do Colo do Útero/genética , Adulto , China , Feminino , Humanos , Pessoa de Meia-Idade
7.
PLoS One ; 15(10): e0236612, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33104714

RESUMO

A precisely balanced activity of canonical Wnt signaling is essential for a number of biological processes and its perturbation leads to developmental defects or diseases. Here, we demonstrate that alternative isoforms of the KDM2A and KDM2B lysine demethylases have the ability to negatively regulate canonical Wnt signaling. These KDM2A and KDM2B isoforms (KDM2A-SF and KDM2B-SF) lack the N-terminal demethylase domain, but they still have the ability to bind to CpG islands in promoters and to interact with their protein partners via their other functional domains. We have observed that KDM2A-SF and KDM2B-SF bind to the promoters of axin 2 and cyclin D1, two canonical Wnt signaling target genes, and repress their activity. Moreover, KDM2A-SF and KDM2B-SF are both able to strongly repress a Wnt-responsive luciferase reporter. The transcriptional repression mediated by KDM2A-SF and KDM2B-SF, but also by KDM2A-LF, is dependent on their DNA binding domain, while the N-terminal demethylase domain is dispensable for this process. Surprisingly, KDM2B-LF is unable to repress both the endogenous promoters and the luciferase reporter. Finally, we show that both KDM2A-SF and KDM2B-SF are able to interact with TCF7L1, one of the transcriptional mediators of canonical Wnt signaling. KDM2A-SF and KDM2B-SF are thus likely to negatively affect the transcription of canonical Wnt signaling target genes by binding to their promoters and by interacting with TCF7L1 and other co-repressors.


Assuntos
Ciclina D1/metabolismo , Proteínas F-Box/metabolismo , Regulação da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Regiões Promotoras Genéticas , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Via de Sinalização Wnt , Ilhas de CpG , Ciclina D1/genética , Proteínas F-Box/genética , Células HEK293 , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Lisina/genética , Lisina/metabolismo , Isoformas de Proteínas , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética
8.
J Biol Chem ; 295(49): 16826-16839, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-32989053

RESUMO

The Wnt/ß-catenin pathway is one of the major pathways that regulates embryonic development, adult homeostasis, and stem cell self-renewal. In this pathway, transcription factors T-cell factor and lymphoid enhancer factor (TCF/LEF) serve as a key switch to repress or activate Wnt target gene transcription by recruiting repressor molecules or interacting with the ß-catenin effector, respectively. It has become evident that the protein stability of the TCF/LEF family members may play a critical role in controlling the activity of the Wnt/ß-catenin signaling pathway. However, factors that regulate the stability of TCF/LEFs remain largely unknown. Here, we report that pVHL binding protein 1 (VBP1) regulates the Wnt/ß-catenin signaling pathway by controlling the stability of TCF/LEFs. Surprisingly, we found that either overexpression or knockdown of VBP1 decreased Wnt/ß-catenin signaling activity in both cultured cells and zebrafish embryos. Mechanistically, VBP1 directly binds to all four TCF/LEF family members and von Hippel-Lindau tumor-suppressor protein (pVHL). Either overexpression or knockdown of VBP1 increases the association between TCF/LEFs and pVHL and then decreases the protein levels of TCF/LEFs via proteasomal degradation. Together, our results provide mechanistic insights into the roles of VBP1 in controlling TCF/LEFs protein stability and regulating Wnt/ß-catenin signaling pathway activity.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Chaperonas Moleculares/metabolismo , Fatores de Transcrição TCF/metabolismo , Via de Sinalização Wnt , Animais , Linhagem Celular , Proliferação de Células , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/genética , Embrião não Mamífero/metabolismo , Humanos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/genética , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição TCF/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Ativação Transcricional , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
9.
Genes (Basel) ; 11(5)2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32403323

RESUMO

Aberrantly activated Wnt signaling causes cellular transformation that can lead to human colorectal cancer. Wnt signaling is mediated by Lymphoid Enhancer Factor/T-Cell Factor (LEF/TCF) DNA-binding factors. Here we investigate whether altered LEF/TCF expression is conserved in human colorectal tumor sample and may potentially be correlated with indicators of cancer progression. We carried out a meta-analysis of carefully selected publicly available gene expression data sets with paired tumor biopsy and adjacent matched normal tissues from colorectal cancer patients. Our meta-analysis confirms that among the four human LEF/TCF genes, LEF1 and TCF7 are preferentially expressed in tumor biopsies, while TCF7L2 and TCF7L1 in normal control tissue. We also confirm positive correlation of LEF1 and TCF7 expression with hallmarks of active Wnt signaling (i.e., AXIN2 and LGR5). We are able to correlate differential LEF/TCF gene expression with distinct transcriptomes associated with cell adhesion, extracellular matrix organization, and Wnt receptor feedback regulation. We demonstrate here in human colorectal tumor sample correlation of altered LEF/TCF gene expression with quantitatively and qualitatively different transcriptomes, suggesting LEF/TCF-specific transcriptional regulation of Wnt target genes relevant for cancer progression and survival. This bioinformatics analysis provides a foundation for future more detailed, functional, and molecular analyses aimed at dissecting such functional differences.


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Fator 1 de Ligação ao Facilitador Linfoide/biossíntese , Proteínas de Neoplasias/biossíntese , Proteína 1 Semelhante ao Fator 7 de Transcrição/biossíntese , Proteína 2 Semelhante ao Fator 7 de Transcrição/biossíntese , Transcriptoma , Via de Sinalização Wnt , Adenocarcinoma/patologia , Proteína Axina/biossíntese , Proteína Axina/genética , Biópsia , Neoplasias Colorretais/patologia , Mineração de Dados , Conjuntos de Dados como Assunto , Progressão da Doença , Retroalimentação Fisiológica , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Proteínas de Neoplasias/genética , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética
10.
Artigo em Inglês | MEDLINE | ID: mdl-31656161

RESUMO

OBJECTIVE: This study aims to discover a potential cytokine biomarker for early diagnosis of thyroid cancer. METHODS: We employed data mining of The Cancer Genome Atlas (TCGA) and experimentally elucidated its mechanistic contributions. The differential expression genes (DEGs) between thyroid cancer and health population were analyzed with TCGA online bioinformatic tools. The relative expression of Bone Morphogenetic Protein 8A (BMP8A) was determined by real-time PCR in ultrasonic diagnosed thyroid cancer both in vivo and in vitro. The serous BMP8A content was quantified with an ELISA kit. Protein levels of BMP8A, OCLN, ZEB1, EZH2 and ß-Actin were analyzed by Western blot. Cell viability was measured by the MTT assay, and anchorage-independent growth was measured by the soft agar colony formation assay. Cell migrative and invasive capacities were interrogated with transwell chamber assays. RESULTS: We identified aberrantly high expression of BMP8A in thyroid cancer, which was associated with unfavorable prognosis and tumor progression. The serous BMP8A was also significantly up-regulated in thyroid cancer patients. Ectopic over-expression of BMP8A remarkably stimulated cell viability and anchorage-independent growth. Meanwhile, the migrative and invasive capacities were greatly increased in response to BMP8A over-expression. Mechanistically, we characterized the positive correlation between BMP8A and TCF7L1, and forced expression of TCF7L1 induced BMP8A expression in TPC-1 cells. CONCLUSION: In summary, we have identified a novel biomarker for early diagnosis in addition to Ultrasound for thyroid cancer, which is subjected to TCF7L1 regulation.


Assuntos
Biomarcadores Tumorais/sangue , Proteínas Morfogenéticas Ósseas/sangue , Progressão da Doença , Neoplasias da Glândula Tireoide/sangue , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Mineração de Dados/métodos , Humanos , Proteína 1 Semelhante ao Fator 7 de Transcrição/sangue
11.
Exp Mol Pathol ; 110: 104293, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31381875

RESUMO

The accurate classification and proper identification of testicular germ cell tumors is imperative for treatment selection and clinical prognosis. Although such distinction can often be achieved by microscopic morphology alone, ancillary tests may at times be needed. T-cell factor 7 L1 (TCF7L1, also known as TCF3), a component of the Wnt signaling pathway, plays important roles in embryonic stem cell self-renewal and lineage specification. Here we examined the immunohistochemical expression and diagnostic utility of TCF7L1 in testicular germ cell tumors. Fifty cases of testicular germ cell tumors were collected, including 23 seminomas, 6 embryonal carcinomas, 1 teratoma, 1 choriocarcinoma, and 19 mixed germ cell tumors. The components of the mixed germ cell tumors were seminoma (n = 3), embryonal carcinoma (n = 18), yolk sac tumor (n = 9), teratoma (n = 15), and choriocarcinoma (n = 4). On immunohistochemistry of TCF7L1, only nuclear staining was considered positive. Staining was graded as negative (<5% of tumor cells stained), minimal (5-25% positive), focal (26-50%), and diffuse (>50%). All non-seminomatous components (n = 54) exhibited distinct nuclear expression of TCF7L1 (54/54; 100%). In contrast, no TCF7L1 expression was detected in the majority of seminomatous tumor component (24/26; 92%). Two seminomas (2/26; 8%) exhibited minimal weak nuclear staining (5% and 10%, respectively) for TCF7L1. In conclusion, TCF7L1, highly expressed in non-seminomatous testicular germ cell tumors, might be used as a marker for diagnosis of testicular germ cell tumors, two therapeutically different entities, for better patient management.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Embrionárias de Células Germinativas/diagnóstico , Seminoma/diagnóstico , Neoplasias Testiculares/diagnóstico , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Adulto , Diagnóstico Diferencial , Humanos , Imuno-Histoquímica , Masculino , Neoplasias Embrionárias de Células Germinativas/metabolismo , Seleção de Pacientes , Seminoma/metabolismo , Neoplasias Testiculares/metabolismo
12.
Stem Cells ; 37(11): 1389-1400, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31322782

RESUMO

Tcf7l1, which is a key effector molecule of the Wnt/ß-catenin signaling pathway, is highly expressed in various cancers, and it promotes tumor growth. In this study, we demonstrated that unlike its tumor-promoting effects in several other types of cancers, Tcf7l1 expression is downregulated in hepatocarcinoma compared with their adjacent nontumor counterparts. Underexpression of Tcf7l1 is correlated with poorer survival. In liver cancer stem cell (CSC) populations, Tcf7l1 expression is downregulated. Ectopic expression of Tcf7l1 attenuates the self-renewal abilities of liver CSCs. Mechanistically, Tcf7l1 regulates the self-renewal abilities of liver CSCs through transcriptional repression of the Nanog gene, and the effect is independent of ß-catenin. Moreover, we found that Tcf7l1 expression is controlled by extracellular insulin-like growth factor (IGF) signaling, and we demonstrated for the first time that IGF signaling stimulates Tcf7l1 phosphorylation and degradation through the mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. Overall, our results provide some new insights into how extracellular signals modulate the self-renewal of liver CSCs and highlight the inhibitory roles of Tcf7l1 in cancer. Stem Cells 2019;37:1389-1400.


Assuntos
Sobrevivência Celular/fisiologia , Fígado/citologia , Fígado/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Somatomedinas/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , beta Catenina/metabolismo , Linhagem Celular , Sobrevivência Celular/genética , Imunoprecipitação da Cromatina , Citometria de Fluxo , Humanos , Imunoensaio , Imuno-Histoquímica , Imunoprecipitação , Técnicas In Vitro , Lentivirus , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Fosforilação , Plasmídeos/genética , Reação em Cadeia da Polimerase em Tempo Real , Somatomedinas/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , beta Catenina/genética
13.
Cell Stem Cell ; 25(2): 210-224.e6, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31104942

RESUMO

Progression through states of pluripotency is required for cells in early mammalian embryos to transition away from heightened self-renewal and toward competency for lineage specification. Here, we use a CRISPR mutagenesis screen in mouse embryonic stem cells (ESCs) to identify unexpected roles for nuclear export and intracellular Ca2+ homeostasis during the exit out of the naive state of pluripotency. Mutation of a plasma membrane Ca2+ pump encoded by Atp2b1 increased intracellular Ca2+ such that it overcame effects of intracellular Ca2+ reduction, which is required for naive exit. Persistent self-renewal of ESCs was supported both in Atp2b1-/-Tcf7l1-/- double-knockout ESCs passaged in defined media alone (no LIF or inhibitors) and in wild-type cells passaged in media containing only calcitonin and a GSK3 inhibitor. These new findings suggest a central role for intracellular Ca2+ in safeguarding naive pluripotency.


Assuntos
Sinalização do Cálcio/fisiologia , Espaço Intracelular/metabolismo , Células-Tronco Embrionárias Murinas/fisiologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Células-Tronco Pluripotentes/fisiologia , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Quinase 3 da Glicogênio Sintase/metabolismo , Homeostase , Camundongos , Camundongos Knockout , Proteínas Nucleares/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética
14.
Acta Biochim Biophys Sin (Shanghai) ; 51(4): 375-385, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30811526

RESUMO

Gastric cancer is one of the most common cancers worldwide and is the third leading cause of cancer-related deaths globally. Although significant progress has been made in the diagnosis and treatment for the cancer, less improvement has been made in overall survival rate. Thus, there is an urgent need for a better understanding of the biological aspects of the cancer. The transcription factor transcription factor 7-like 1 (TCF7L1) is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in gastric cancer has seldom been discussed. In the present study, by using the Cancer Genome Atlas dataset analysis, we demonstrated that patients with higher expression of TCF7L1 could be used to reflect prognosis. An examination of the mechanisms demonstrated that TCF7L1 could positively regulate antioxidant response in gastric cancer cells by positively regulating Keap1/NRF2 [Kelch-like ECH-associated protein 1/nuclear factor (erythroid-derived 2)-like 2] pathway. Collectively, our data demonstrated that TCF7L1 is a novel marker for predicting overall survival of gastric cancer and provided the possible underlying molecular mechanism.


Assuntos
Proliferação de Células/genética , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fator 2 Relacionado a NF-E2/genética , Neoplasias Gástricas/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Antioxidantes/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Glicólise/genética , Humanos , Estimativa de Kaplan-Meier , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Masculino , Pessoa de Meia-Idade , Fator 2 Relacionado a NF-E2/metabolismo , Prognóstico , Transdução de Sinais/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo
15.
Cell Rep ; 25(3): 571-584.e5, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30332639

RESUMO

Wnt/ß-catenin signaling is necessary for lymphatic vascular development. Oscillatory shear stress (OSS) enhances Wnt/ß-catenin signaling in cultured lymphatic endothelial cells (LECs) to induce expression of the lymphedema-associated transcription factors GATA2 and FOXC2. However, the mechanisms by which OSS regulates Wnt/ß-catenin signaling and GATA2 and FOXC2 expression are unknown. We show that OSS activates autocrine Wnt/ß-catenin signaling in LECs in vitro. Tissue-specific deletion of Wntless, which is required for the secretion of Wnt ligands, reveals that LECs and vascular smooth muscle cells are complementary sources of Wnt ligands that regulate lymphatic vascular development in vivo. Further, the LEC master transcription factor PROX1 forms a complex with ß-catenin and the TCF/LEF transcription factor TCF7L1 to enhance Wnt/ß-catenin signaling and promote FOXC2 and GATA2 expression in LECs. Thus, our work defines Wnt sources, reveals that PROX1 directs cell fate by acting as a Wnt signaling component, and dissects the mechanisms of PROX1 and Wnt synergy.


Assuntos
Células Endoteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Músculo Liso Vascular/citologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA2/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Via de Sinalização Wnt
16.
Mol Cell ; 72(1): 37-47.e4, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30197296

RESUMO

Adenomatous polyposis coli (APC) and Axin are core components of the ß-catenin destruction complex. How APC's function is regulated and whether Wnt signaling influences the direct APC-Axin interaction to inhibit the ß-catenin destruction complex is not clear. Through a CRISPR screen of ß-catenin stability, we have identified ICAT, a polypeptide previously known to block ß-catenin-TCF interaction, as a natural inhibitor of APC. ICAT blocks ß-catenin-APC interaction and prevents ß-catenin-mediated APC-Axin interaction, enhancing stabilization of ß-catenin in cells harboring truncated APC or stimulated with Wnt, but not in cells deprived of a Wnt signal. Using ICAT as a tool to disengage ß-catenin-mediated APC-Axin interaction, we demonstrate that Wnt quickly inhibits the direct interaction between APC and Axin. Our study highlights an important scaffolding function of ß-catenin in the assembly of the destruction complex and suggests Wnt-inhibited APC-Axin interaction as a mechanism of Wnt-dependent inhibition of the destruction complex.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Domínios e Motivos de Interação entre Proteínas/genética , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteína da Polipose Adenomatosa do Colo/antagonistas & inibidores , Proteína Axina/genética , Humanos , Estabilidade Proteica , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Via de Sinalização Wnt/genética
17.
Am J Physiol Cell Physiol ; 314(6): C712-C720, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29513567

RESUMO

Although long noncoding RNAs (lncRNAs) are emerging as new modulators in the fate decision of pluripotent stem cells, the functions of specific lncRNAs remain unclear. Here, we found that telomeric RNA (TERRA or TelRNA), one type of lncRNAs, is highly expressed in mouse embryonic stem cells (mESCs) but declines significantly upon differentiation. TERRA is induced by the Wnt/ß-catenin signaling pathway and can reproduce its self-renewal-promoting effect when overexpressed. Further studies revealed that T cell factor 3 ( TCF3) is a potential downstream target of TERRA and mediates the effect of TERRA in mESC maintenance. TERRA inhibits TCF3 transcription, while enforced TCF3 expression abrogates the undifferentiated state of mESCs supported by TERRA. Accordingly, the transcripts of the pluripotency genes Esrrb, Tfcp2l1, and Klf2, repressed by TCF3 in mESCs, are increased in TERRA-overexpressing cells. Our study therefore highlights the important role of TERRA in mESC maintenance and also uncovers a mechanism by which TERRA promotes self-renewal. These data will expand our understanding of the pluripotent regulatory network of ESCs.


Assuntos
Autorrenovação Celular , Células-Tronco Embrionárias Murinas/metabolismo , RNA não Traduzido/metabolismo , Telômero/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Autorrenovação Celular/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos da Linhagem 129 , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , RNA não Traduzido/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Telômero/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Transcrição Gênica , Via de Sinalização Wnt
18.
Mol Cell Biol ; 37(18)2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28674186

RESUMO

Transforming growth factor ß (TGF-ß)-induced migration of triple-negative breast cancer (TNBC) cells is dependent on nuclear export of the orphan receptor NR4A1, which plays a role in proteasome-dependent degradation of SMAD7. In this study, we show that TGF-ß induces p38α (mitogen-activated protein kinase 14 [MAPK14]), which in turn phosphorylates NR4A1, resulting in nuclear export of the receptor. TGF-ß/p38α and NR4A1 also play essential roles in the induction of epithelial-to-mesenchymal transition (EMT) and induction of ß-catenin in TNBC cells, and these TGF-ß-induced responses and nuclear export of NR4A1 are blocked by NR4A1 antagonists, the p38 inhibitor SB202190, and kinase-dead [p38(KD)] and dominant-negative [p38(DN)] forms of p38α. Inhibition of NR4A1 nuclear export results in nuclear export of TGF-ß-induced ß-catenin, which then undergoes proteasome-dependent degradation. TGF-ß-induced ß-catenin also regulates NR4A1 expression through formation of the ß-catenin-TCF-3/TCF-4/LEF-1 complex on the NR4A1 promoter. Thus, TGF-ß-induced nuclear export of NR4A1 in TNBC cells plays an essential role in cell migration, SMAD7 degradation, EMT, and induction of ß-catenin, and all of these pathways are inhibited by bis-indole-derived NR4A1 antagonists that inhibit nuclear export of the receptor and thereby block TGF-ß-induced migration and EMT.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fator de Crescimento Transformador beta/genética , Neoplasias de Mama Triplo Negativas/patologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Imidazóis/farmacologia , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/biossíntese , Complexos Multiproteicos/biossíntese , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/antagonistas & inibidores , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Fosforilação , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína Smad7/metabolismo , Fator de Transcrição 4 , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo
19.
Elife ; 62017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28467300

RESUMO

The transcription factor TCF7L1 is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in skin tumorigenesis has not yet been defined. Here we document TCF7L1 upregulation in skin squamous cell carcinoma (SCC) and demonstrate that TCF7L1 overexpression increases tumor incidence, tumor multiplicity, and malignant progression in the chemically induced mouse model of skin SCC. Additionally, we show that downregulation of TCF7L1 and its paralogue TCF7L2 reduces tumor growth in a xenograft model of human skin SCC. Using separation-of-function mutants, we show that TCF7L1 promotes tumor growth, enhances cell migration, and overrides oncogenic RAS-induced senescence independently of its interaction with ß-catenin. Through transcriptome profiling and combined gain- and loss-of-function studies, we identified LCN2 as a major downstream effector of TCF7L1 that drives tumor growth. Our findings establish a tumor-promoting role for TCF7L1 in skin and elucidate the mechanisms underlying its tumorigenic capacity.


Assuntos
Carcinogênese , Carcinoma de Células Escamosas/fisiopatologia , Lipocalina-2/metabolismo , Neoplasias Cutâneas/fisiopatologia , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , beta Catenina/metabolismo , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Xenoenxertos , Humanos , Camundongos
20.
Biochem Biophys Res Commun ; 487(3): 716-722, 2017 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-28450117

RESUMO

The T-cell factor/Lymphoid enhancer factor (TCF/LEF; hereafter TCF) family of transcription factors are critical regulators of colorectal cancer (CRC) cell growth. Of the four TCF family members, TCF7L1 functions predominantly as a repressor of gene expression. Few studies have addressed the role of TCF7L1 in CRC and only a handful of target genes regulated by this repressor are known. By silencing TCF7L1 expression in HCT116 cells, we show that it promotes cell proliferation and tumorigenesis in vivo by driving cell cycle progression. Microarray analysis of transcripts differentially expressed in control and TCF7L1-silenced CRC cells identified genes that control cell cycle kinetics and cancer pathways. Among these, expression of the Wnt antagonist DICKKOPF4 (DKK4) was upregulated when TCF7L1 levels were reduced. We found that TCF7L1 recruits the C-terminal binding protein (CtBP) and histone deacetylase 1 (HDAC1) to the DKK4 promoter to repress DKK4 gene expression. In the absence of TCF7L1, TCF7L2 and ß-catenin occupancy at the DKK4 promoter is stimulated and DKK4 expression is increased. These findings uncover a critical role for TCF7L1 in repressing DKK4 gene expression to promote the oncogenic potential of CRCs.


Assuntos
Oxirredutases do Álcool/metabolismo , Neoplasias Colorretais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desacetilase 1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA