Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Cell Int ; 21(1): 392, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34289835

RESUMO

BACKGROUND: Long noncoding RNAs (lncRNAs) have emerged as crucial regulators in various cancers. However, the functional roles of most lncRNA in papillary thyroid cancer (PTC) are not detailly understood. This study aims to investigate the biological function and molecular mechanism of lncRNA Fer-1 like family member 4 (FER1L4) in PTC. METHODS: The expression of FER1L4 in PTC was determined via operating quantitative real-time PCR assays. Meanwhile, the clinical significance of FER1L4 in patients with PTC was described. The biological functions of FER1L4 on PTC cells were evaluated by gain and loss of function experiments. Moreover, animal experiments were performed to reveal the effect on tumor growth. Subcellular distribution of FER1L4 was determined by fluorescence in situ hybridization and subcellular localization assays. Luciferase reporter assay and RNA immunoprecipitation assay were applied to define the relationship between FER1L4, miR-612, and Cadherin 4 (CDH4). RESULTS: Upregulated expression of FER1L4 in PTC tissues was positively correlated with lymph node metastasis (P = 0.020), extrathyroidal extension (P = 0.013) and advanced TNM stages (P = 0.013). In addition, knockdown of FER1L4 suppressed PTC cell proliferation, migration, and invasion, whereas ectopic expression of FER1L4 inversely promoted these processes. Mechanistically, FER1L4 could competitively bind with miR-612 to prevent the degradation of its target gene CDH4. This condition was further confirmed in the rescue assays. CONCLUSIONS: This study first demonstrates FER1L4 plays an oncogenic role in PTC via a FER1L4-miR-612-CDH4 axis and may provide new therapeutic and diagnostic targets for PTC.

2.
Mol Biol Rep ; 48(5): 4581-4589, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34132945

RESUMO

Colorectal cancer (CRC) is a major life-threatening malignancy. Studies demonstrated the lncRNA fer-1 like family member 4 (FER1L4) was downregulated in different cancers and its expression was positively correlated with the retinoblastoma 1 (RB1) mRNA in a competing endogenous RNAs network. We investigated expression levels of FER1L4 and RB1 in patients with colorectal cancer. 50 paired colorectal tumors and non-tumor marginal tissues, 30 paired adenomatous colorectal polyps (ACPs) and matched adjacent normal tissues were obtained from the patients. Total RNA was extracted from the samples and cDNAs were synthesized. Their expression was quantified by qRT-PCR. Correlation between FER1L4 and RB1 expression levels was analyzed by Pearson correlation test. Finally, ROC curve analysis was used to evaluate their biomarker potency. We observed significant downregulation of FER1L4, but upregulation of RB1 in the colorectal tumors compared with non-tumor and the polyp tissues. However, RB1 expression was positively correlated with FER1L4 expression both in the tumor and polyp samples. ROC curve analysis showed both FER1L4 and RB1 expression levels could discriminate tumor from non-tumor and tumor from polyp samples. None of the clinicopathological characteristics of patients were associated with FER1L4 or RB1 expression levels. Despite the downregulation of FER1L4 and upregulation of RB1 in tumors compared with non-tumor tissues, the expression of RB1 was positively correlated with the expression of FER1L4 in the colorectal tumor as well as in the polyp tissues. FER1L4 expression level might be considered as a potential biomarker for colorectal cancer development.


Assuntos
Polipose Adenomatosa do Colo/genética , Neoplasias Colorretais/genética , RNA Longo não Codificante/genética , Proteínas de Ligação a Retinoblastoma/genética , Ubiquitina-Proteína Ligases/genética , Polipose Adenomatosa do Colo/patologia , Idoso , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Pessoa de Meia-Idade
3.
J Cell Physiol ; 235(2): 1795-1807, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31332783

RESUMO

The function of Fer-1 like family member 4 (FER1L4) in human pan-cancer is unknown. Expression of FER1L4 in tumor tissues and nontumor tissues, upstream regulation of FER1L4, and the relationship between its expression with prognosis and chemoresistance were examined by The Cancer Genome Atlas and Gene Expression Omnibus databases. Next, these results were validated in breast tumor and paired nontumor tissues in our cohort. FER1L4 expression is higher in tumor tissues compared with the adjacent nontumor tissues. High FER1L4 expression is associated with worse disease outcomes. Hypomethylation and H3K4me3 accumulation in FER1L4 promoter locus activate its transcriptional expression. Moreover, FER1L4 may trigger chemoresistance in human cancer. Gene Ontology enrichment analysis revealed that FER1L4 may be involved in processes associated with tumorigenesis. These results indicated that FER1L4 may act as an oncogenic driver and it may be a potential therapy target in human cancer.


Assuntos
Carcinogênese/genética , Neoplasias/genética , Oncogenes/genética , RNA Longo não Codificante/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos
4.
Cancer Cell Int ; 20: 64, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32140077

RESUMO

BACKGROUND: Dysregulation of long non-coding RNAs (lncRNAs) is involved in development of prostate cancer. However, the molecular mechanisms of many lncRNAs in prostate cancer have not been studied yet. METHODS: The lncRNA Fer-1-like protein 4 (FER1L4) expression was explored in prostate tumors and normal prostate tissues by RT-qPCR and bioinformatic analysis. Overexpression of FER1L4 was performed to evaluate its role in prostate cancer cell proliferation and survival. The molecular mechanism of FER1L4 was investigated by dual luciferase reporter assay, RNA pull down assay, western blotting and RT-qPCR. RESULTS: It was found that FER1L4 was lower in prostate cancer tissues than normal tissues. Higher expression of FER1L4 was associated with prostate cancer tissues of early stage (AJCC stage I/II). Overexpression of FER1L4 inhibited cell proliferation and promoted cell apoptosis in prostate cancer cells. Bioinformatic analysis, RT-qPCR, RNA pull down assay and dual luciferase assay showed that FER1L4 upregulated F-box/WD repeat-containing protein 7 (FBXW7) tumor suppressor via sponging miR-92a-3p. Silencing of FBXW7 reversed the cell phenotypes caused by FER1L4 overexpression in prostate cancer cells. CONCLUSION: The data demonstrated that FER1L4, a downregulated lncRNA in prostate cancer, was pivotal for cell proliferation and survival of prostate cancer. The study provided new sights into understanding of the signaling network in prostate cancer and implied that FER1L4 might be a biomarker for patients with prostate cancer.

5.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33228208

RESUMO

The reciprocal interactions between cancer cells and the quiescent fibroblasts leading to the activation of cancer-associated fibroblasts (CAFs) serve an important role in cancer progression. Here, we investigated the activation of transcription factors (TFs) in prostate fibroblasts (WPMY cell line) co-cultured with normal prostate or tumorous cells (RWPE1 and RWPE2 cell lines, respectively). After indirect co-cultures, we performed mRNA-seq and predicted TF activity using mRNA expression profiles with the Systems EPigenomics Inference of Regulatory Activity (SEPIRA) package and the GTEx and mRNA-seq data of 483 cultured fibroblasts. The initial differential expression analysis between time points and experimental conditions showed that co-culture with normal epithelial cells mainly promotes an inflammatory response in fibroblasts, whereas with the cancerous epithelial, it stimulates transformation by changing the expression of the genes associated with microfilaments. TF activity analysis revealed only one positively regulated TF in the RWPE1 co-culture alone, while we observed dysregulation of 45 TFs (7 decreased activity and 38 increased activity) uniquely in co-culture with RWPE2. Pathway analysis showed that these 45 dysregulated TFs in fibroblasts co-cultured with RWPE2 cells may be associated with the RUNX1 and PTEN pathways. Moreover, we showed that observed dysregulation could be associated with FER1L4 expression. We conclude that phenotypic changes in fibroblast responses to co-culturing with cancer epithelium result from orchestrated dysregulation of signaling pathways that favor their transformation and motility rather than proinflammatory status. This dysregulation can be observed both at the TF and transcriptome levels.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição/genética , Fibroblastos Associados a Câncer/patologia , Comunicação Celular , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Técnicas de Cocultura , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Ontologia Genética , Humanos , Masculino , Anotação de Sequência Molecular , PTEN Fosfo-Hidrolase/metabolismo , Próstata/metabolismo , Próstata/patologia , Transdução de Sinais , Fatores de Transcrição/classificação , Fatores de Transcrição/metabolismo , Transcriptoma
6.
Rep Pract Oncol Radiother ; 25(5): 808-819, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32884453

RESUMO

miR-18a is a member of primary transcript called miR-17-92a (C13orf25 or MIR17HG) which also contains five other miRNAs: miR-17, miR-19a, miR-20a, miR-19b and miR-92a. This cluster as a whole shows specific characteristics, where miR-18a seems to be unique. In contrast to the other members, the expression of miR-18a is additionally controlled and probably functions as its own internal controller of the cluster. miR-18a regulates many genes involved in proliferation, cell cycle, apoptosis, response to different kinds of stress, autophagy and differentiation. The disturbances of miR-18a expression are observed in cancer as well as in different diseases or pathological states. The miR-17-92a cluster is commonly described as oncogenic and it is known as 'oncomiR-1', but this statement is a simplification because miR-18a can act both as an oncogene and a suppressor. In this review we summarize the current knowledge about miR-18a focusing on its regulation, role in cancer biology and utility as a potential biomarker.

7.
J Cell Mol Med ; 23(5): 3224-3233, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30887657

RESUMO

Long non-coding RNAs have recently become a key regulatory factor for cancers, whereas FER1L4, a newly discovered long non-coding RNA, has been mostly studied in gastric carcinoma and colon cancer cases. The functions and molecular mechanism of FER1L4 have been rarely reported in glioma malignant phenotypes. In this study, it was found that the expression of LncRNA FER1L4 is upregulated in high-grade gliomas than in low-grade cases and that a high expression of LncRNA FER1L4 predicts poor prognosis of gliomas. Meanwhile, in vitro study suggests that expression of FER1L4 with SiRNA knockdown obviously suppresses cell cycle and proliferation. It is further demonstrated by experiments that the FER1L4 knockdown suppresses growth of in vivo glioma. Besides, it is found in our study that LncRNA FER1L4 expression is positively correlated with E2F1 mRNA expression. After knockdown of FER1L4 expression, E2F1 expression is significantly down-regulated, whereas the expression of miR-372 is significantly up-regulated; the up-regulation of miR-372 leads to significant down-regulation of FER1L4 and E2F1 expression. In addition, it is also found that FER1L4 can be used as competitive endogenous RNA to interact or bind with miR-371 and thereby up-regulate E2F1, thus promoting the cycle and proliferation of glioma cells. It may be one of the molecular mechanisms in which FER1L4 plays its oncogene-like role in gliomas.


Assuntos
Fator de Transcrição E2F1/genética , Glioma/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/patologia , Humanos
8.
J Cell Biochem ; 120(5): 7581-7589, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30444026

RESUMO

To determine how the lncRNA FER1L4 in ovarian cancer cells influences paclitaxel (PTX) resistance, we examined the expression level of FER1L4 in human ovarian epithelial cell lines IOSE80 and HOSEpiC and human ovarian cancer cell lines OVCAR-3, Caov-3, and SKOV3 through RNA isolation and quantitative polymerase chain reaction (qRT-PCR). SKOV3 cell lines were treated with PTX. The cell survival rate and apoptosis rate of SKOV3 and SKOV3-PR at different PTX dose levels were evaluated. Next, qRT-PCR was performed to detect the expression of FER1L4 in SKOV3 and SKOV3-PR cell lines. SKOV3-PR cell lines were transfected with pcDNA3.1 as the control group (SKOV3-PR/pcDNA3.1) or pcDNA3.1-FER1L4 to upregulate the expression level of FER1L4 (SKOV3-PR/pcDNA3.1-FER1L4). The level of cell survival, apoptosis, and colony formation were compared between the two groups using MTT, flow cytometry analysis, and colony formation assay. To reveal the molecular mechanism, we measured the relative protein phosphorylation level of ERK and MAPK in SKOV3, SKOV3-PR, SKOV3-PR/pcDNA3.1, and SKOV3-PR/pcDNA3.1-FER1L4 groups using an enzyme-linked immunosorbent assay. The effects of SB203580 (a p38 MAPK inhibitor) on PTX were also investigated to reveal the function of the MAPK pathway on the PTX tolerance of SKOV3. In comparison with normal ovarian epithelial cells, FER1L4 was downregulated. The FER1L4 level was decreased in human ovarian cancer cells with drug resistance than in common ovarian cancer cells. The upregulation of FER1L4 could promote the PTX sensitivity of ovarian cancer cells. The increased level of FER1L4 could suppress the PTX resistance of ovarian cancer cells through the inhibition of the MAPK signaling pathway.

9.
J Cell Biochem ; 120(4): 6781-6788, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30382631

RESUMO

BACKGROUND AND OBJECTIVES: This study aimed to investigate the potential function of FER1L4 in the progression of hepatocellular carcinoma and uncover its underlying molecular mechanism. METHODS: In the current study, quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to determine the expression profile of FER1L4 in normal liver tissues and hepatocellular carcinoma tissues of human, as well as hepatocellular carcinoma (HCC) cell lines including HL-7702[L-02], HepG-2, Hep3b, and SMMC-7721. Then, HepG-2 cells were transfected with pcDNA3.1-FER1L4 (pcDNA3.1-empty as negative control) for gain-of-function analysis, followed with cell functional abnormality tests. Specifically, colony formation analysis and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide experiment were taken advantage to measure the cell proliferation, while cell migration and invasion were evaluated by wound healing assay and transwell experiment respectively. Additionally, cell apoptosis was detected by flow cytometry. Moreover, the effect of FER1L4 on PI3K/AKT signal pathway activation was investigated through analyzing phosphorylation of related proteins, p-AKT/AKT and p-PI3K/PI3K, via Western blot assay. RESULTS: Downregulation of FER1L4 in hepatocellular carcinoma tissues and cells was demonstrated by qRT-PCR analysis. Besides, FER1L4 overexpression evidently attenuated the cell proliferation, migration and invasion, but prompted cell apoptosis. Importantly, Western blot assays revealed that PII3K/AKT signal pathway were involved in mediating the progression regulation role of FER1L4 in HCC cells. CONCLUSIONS: Our study suggested that FER1L4 might alleviate progression of hepatocellular carcinoma via blocking PI3K/AKT pathway, which encourages a better understanding of the pathogenesis of HCC and may provide a novel potential therapeutic target for clinical treatment.


Assuntos
Carcinoma Hepatocelular/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Hepáticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Regulação para Cima/genética , Apoptose/genética , Carcinoma Hepatocelular/patologia , Regulação Neoplásica da Expressão Gênica/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Invasividade Neoplásica/genética , Fosforilação/genética , RNA Longo não Codificante/genética , Transdução de Sinais/genética , Transfecção
10.
Traffic ; 17(3): 245-66, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707827

RESUMO

Ferlins are a family of transmembrane-anchored vesicle fusion proteins uniquely characterized by 5-7 tandem cytoplasmic C2 domains, Ca(2+)-regulated phospholipid-binding domains that regulate vesicle fusion in the synaptotagmin family. In humans, dysferlin mutations cause limb-girdle muscular dystrophy type 2B (LGMD2B) due to defective Ca(2+)-dependent, vesicle-mediated membrane repair and otoferlin mutations cause non-syndromic deafness due to defective Ca(2+)-triggered auditory neurotransmission. In this study, we describe the tissue-specific expression, subcellular localization and endocytic trafficking of the ferlin family. Studies of endosomal transit together with 3D-structured illumination microscopy reveals dysferlin and myoferlin are abundantly expressed at the PM and cycle to Rab7-positive late endosomes, supporting potential roles in the late-endosomal pathway. In contrast, Fer1L6 shows concentrated localization to a specific compartment of the trans-Golgi/recycling endosome, cycling rapidly between this compartment and the PM via Rab11 recycling endosomes. Otoferlin also shows trans-Golgi to PM cycling, with very low levels of PM otoferlin suggesting either brief PM residence, or rare incorporation of otoferlin molecules into the PM. Thus, type-I and type-II ferlins segregate as PM/late-endosomal or trans-Golgi/recycling ferlins, consistent with different ferlins mediating vesicle fusion events in specific subcellular locations.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Membrana Celular/metabolismo , Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Rede trans-Golgi/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Células COS , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/genética , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Especificidade de Órgãos , Pâncreas/metabolismo , Transporte Proteico , Proteínas rab de Ligação ao GTP/metabolismo
11.
Biochem Biophys Res Commun ; 478(2): 507-12, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27381864

RESUMO

BACKGROUND: Dysregulated long non-coding RNAs (lncRNAs) might exert key roles in pathways associated with endometrial carcinoma (EC) development. This study aims to investigate the new role of lncRNA FER1L4 in EC pathogenesis due to its correlation with phosphatase and tensin homolog (PTEN), one important indicator of EC progression. METHODS: Real time PCR was performed to detect the expression of FER1L4 in thirty paired EC samples and two EC cell lines. Plasmid containing FER1L4 was transfected into HEC-50 cells with a relative lower level of FER1L4 expression, followed which PTEN expression and Akt phosphorylation were measured by western blotting. Cell proliferation was analyzed through MTT and colony-formation assays, while cell cycle and apoptosis were determined by flow cytometry. RESULTS: FER1L4 showed significantly downregulation in EC tissues compared to control, which was positively correlated with decreased PTEN expression. Moreover, FER1L4 could promote PTEN expression and inhibit Akt phosphorylation. Additionally, a significant decrease of cell proliferation was observed in FER1L4 overexpressing cells, along with cell cycle arrest at G0/G1 phase and increased proportion of apoptotic cells. CONCLUSION: FER1L4 not only showed downregulation in EC tissues and cells, but also regulated PTEN expression and Akt signaling, which might contribute to its inhibition on cell proliferation. This study might provide a new potential therapeutic target for EC treatment.


Assuntos
Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Endométrio/patologia , Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/genética , RNA Longo não Codificante/genética , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos
12.
Cancer Sci ; 106(10): 1323-32, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26224446

RESUMO

Novel long non-coding RNA Fer-1-like protein 4 (FER1L4) has been confirmed to play crucial regulatory roles in tumor progression. It exerts an impact on tumor suppression and functions as a competing endogenous RNA (ceRNA) by sponging miR-106a-5p in gastric cancer. However, its clinical significance in colon cancer is completely unknown. The aim of the present study was to annotate the role of FER1L4 and its clinical value in colon cancer. The results showed the aberrant expression of FER1L4 and miR-106a-5p in colon cancer tissues. In addition, significant negative correlation between FER1L4 and miR-106a-5p expression levels was observed. Among the colon cancer cell lines, FER1L4 levels were relatively lower, with concurrent high levels of miR-106a-5p. Restoration of FER1L4 decreased the expression of miR-106a-5p, and had a significant influence on colon cancer cell proliferation, migration and invasion. The FER1L4 expression was correlated with depth of tumor invasion, lymph node metastasis, vascular invasion and clinical stage. Moreover, striking differences in overall survival and disease-free survival were observed for the cases with both low FER1L4 expression and high miR-106a-5p expression compared with cases with high FER1L4 expression and low miR-106a-5p expression. Circulating FER1L4 and miR-106a-5p levels were decreased and increased, respectively, in colon cancer patients after surgery. Our findings indicated that FER1L4 could exert a tumor suppressive impact on colon cancer, which at least, in part, through suppressing miR-106a-5p expression, and depletion of FER1L4, alone or combined with overexpression of miR-106a-5p, is predictive of poor prognosis in colon cancer and may play a crucial role in cancer prevention and treatment.


Assuntos
Transformação Celular Neoplásica/patologia , Neoplasias do Colo/patologia , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Neoplasias do Colo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Metástase Linfática , Masculino , MicroRNAs/sangue , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , RNA Longo não Codificante/sangue , RNA Longo não Codificante/genética
13.
Pathol Res Pract ; 257: 155291, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38643553

RESUMO

Because long non-coding RNAs (lncRNAs) can affect several interconnected processes, its value as a predictive marker for gastric cancer has been demonstrated. Coumarin - a natural compound known to contain some beneficial antitumor qualities - was tested for its effects on AGS gastric cancer cells. In this study, we investigated the expression level of selected cellular lncRNAs (BANCR, MALAT1 and FER1L4) and their target genes (PTEN, p-PI3K and p-AKT) in coumarin-treated AGS cell line. The expressions of the three lncRNAs: BANCR, MALAT1 and FER1L4, as well as their specified targets, PTEN, PI3K and AKT, were measured by qRT-PCR. To gauge the impact of coumarin on the AGS cells, a MTT assay was utilized. A Western blot has been employed to assess variations in PTEN, p-PI3K, and p-AKT expression. The experiment's results showed that AGS viability diminished with increasing doses of coumarin. Compared to the control cells, the cells exposed to coumarin had showed reduced levels of mRNAs which are known targets of the lncRNA BANCR. At the same time, levels of lncRNAs MALAT1 and FER1L4 within coumarin group have been higher comparing to those within control group. Additionally, the Western blot analysis revealed that the coumarin-treated cells expressed lower levels of p-PI3K, PTEN as well as p-AKT compared to control group. This information points to coumarin being a possible option in a treatment regimen for gastric cancer due to its ability to affect lncRNAs and the molecules they target.


Assuntos
Cumarínicos , RNA Longo não Codificante , Neoplasias Gástricas , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Humanos , Cumarínicos/farmacologia , Linhagem Celular Tumoral , Neoplasias Gástricas/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo
14.
Biol Direct ; 19(1): 57, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39039611

RESUMO

Laryngeal carcinoma (LC) is a common cancer of the respiratory tract. This study aims to investigate the role of RNA-binding motif protein 15 (RBM15) in the cisplatin (DDP) resistance of LC cells. LC-DDP-resistant cells were constructed. RBM15, lysine-specific demethylase 5B (KDM5B), lncRNA Fer-1 like family member 4 (FER1L4), lncRNA KCNQ1 overlapping transcript 1 (KCNQ1OT1), glutathione peroxidase 4 (GPX4), and Acyl-CoA synthetase long-chain family (ACSL4) was examined. Cell viability, IC50, and proliferation were assessed after RBM15 downregulation. The enrichment of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) and N6-methyladenosine (m6A) on KDM5B was analyzed. KDM5B mRNA stability was measured after actinomycin D treatment. A tumor xenograft assay was conducted to verify the role of RBM15 in LC. Results showed that RBM15 was upregulated in LC and its knockdown decreased IC50, cell viability, proliferation, glutathione, and upregulated iron ion content, ROS, malondialdehyde, ACSL4, and ferroptosis. Mechanistically, RBM15 improved KDM5B stability in an IGF2BP3-dependent manner, resulting in FER1L4 downregulation and GPX4 upregulation. KDM5B increased KCNQ1OT1 and inhibited ACSL4. KDM5B/KCNQ1OT1 overexpression or FER1L4 knockdown promoted DDP resistance in LC by inhibiting ferroptosis. In conclusion, RBM15 promoted KDM5B expression, and KDM5B upregulation inhibited ferroptosis and promoted DDP resistance in LC by downregulating FER1L4 and upregulating GPX4, as well as by upregulating KCNQ1OT1 and inhibiting ACSL4. Silencing RBM15 inhibited tumor growth in vivo.


Assuntos
Cisplatino , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética , Ferroptose , Neoplasias Laríngeas , Proteínas de Ligação a RNA , Ferroptose/genética , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular Tumoral , Camundongos , Animais , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Camundongos Nus , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/efeitos dos fármacos , Antineoplásicos/farmacologia , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo
15.
Asian Pac J Cancer Prev ; 24(7): 2271-2277, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37505756

RESUMO

BACKGROUND: Numerous studies have probed the deregulation of the long noncoding RNA AB073614 and FER1L4, which have been discovered in a variety of cancers. However, the precise expression pattern of these lncRNAs and their clinical implications in acute myeloid leukemia (AML) remain elusive. Considering the involvement of the PI3K axis in AML pathogenesis, an investigation into the expression of AB073614 and FER1L4 targets of this pathway has been proposed, aiming to elucidate a potential mechanism underlying AML development. METHODS: The expression levels of lncRNA AB073614 and FER1L4 were assessed in 30 newly diagnosed AML patients and 12 healthy individuals using quantitative reverse transcription-polymerase chain reaction techniques. A statistical analysis was conducted to determine the association of AB073614 and FER1L4 expression levels with clinicopathological features. RESULTS: A significant upregulation of AB073614 was observed in AML patients compared to the control group (p < 0.05). Moreover, a notable increase in AB073614 expression levels coincided with a significant reduction in FER1L4 expression levels in AML samples (p < 0.05). The diagnostic value of these lncRNAs was validated using the receiver operating characteristic (ROC) curve and area under the curve (AUC) calculations. Sensitivity values of AB073614 and FER1L4 gene expression were 96.7% and 100%, respectively, using cut-off relative quantification of 1.045 and 0.770. Additionally, specificity values were observed to be 100%. CONCLUSIONS: The present study indicates that AB073614 and FER1L4 might serve as prognosis biomarkers in AML patients. However, further detailed examinations in this field are warranted. It is proposed that the likely mechanism of imbalanced PI3K and PTEN activity, triggered by the deregulation of AB073614 and FER1L4, may have a crucial role in AML pathogenesis. Any component of this pathway could potentially serve as a new target for more insightful treatment approaches.


Assuntos
Leucemia Mieloide Aguda , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Linhagem Celular Tumoral , Regulação para Cima , Leucemia Mieloide Aguda/genética , Fosfatidilinositol 3-Quinases/genética , Prognóstico
16.
Am J Transl Res ; 14(4): 2256-2266, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35559379

RESUMO

Neural stem cells (NSCs) may offer beneficeial and promising adjuncts for treatment of neurological diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and spinal cord injuries. Previous studies showed that LncRNA FER1L4 plays crucial roles in many biological procedures such as invasion, metabolism, apoptosis, and stem cell differentiation. However, the role of FER1L4 in differentiation and growth of NSCs remains unknown. In the present research, we noted that FER1L4 is upregulated in NSCs induced with TNFα. Ectopic expression of FER1L4 suppresses NSCs proliferation and induces NSCs differentiated into neurons and astrocytes. Using Starbase online software, we identified that FER1L4 is one potential target gene of miR-874-3p. Ectopic expression of FER1L4 decreases miR-874-3p expression in NSCs. We identified Ascl2 is one target gene for miR-874-3p. Overexpression of FER1L4 enhances Ascl2 expression in NSCs. Furthermore, we proved that FER1L4 modulates the proliferation and differentiation of NSCs via regulating Ascl2.

17.
Curr Pharm Des ; 28(16): 1334-1341, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35331091

RESUMO

BACKGROUND: FER-1 family member 4 (FER1L4), a 6.7 kb lncRNA located at 20q11.22, plays an important biological function in a variety of tumor diseases. The purpose of this review is to clarify the pathophysiological mechanism and potential biological function of FER1L4 in different tumors. METHODS: By searching the relevant literature in PubMed, the specific pathophysiological mechanism of FER1L4 in different tumors was summarized. RESULTS: LncRNA FER1L4 is one of the key factors in tumorigenesis and is abnormally down-regulated in many tumors, including osteosarcoma, lung cancer, laryngeal squamous cell carcinoma, laryngeal cancer, colorectal cancer, ovarian cancer, prostate cancer, esophageal cancer, gastric cancer, endometrial cancer, osteoarthritis, rheumatoid arthritis, and so on. However, FER1L4 is downregulated in breast cancer, glioma, oral squamous cell carcinoma, renal clear cell carcinoma, and periodontitis, and plays a protective role in orthodontic teeth. In addition, as a tumor suppressor gene or oncogene, FER1L4 affects tumor proliferation, invasion, migration, and apoptosis. CONCLUSION: LncRNA FER1L4 has a good application prospect in the treatment and diagnosis of various tumors.


Assuntos
Neoplasias , RNA Longo não Codificante , Neoplasias Ósseas/patologia , Carcinoma de Células Escamosas/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Bucais/genética , Neoplasias/genética , RNA Longo não Codificante/genética
18.
Onco Targets Ther ; 14: 795-806, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33568918

RESUMO

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a common cancer especially young people in the world. The long non-coding RNA Fer-1-like protein 4 (FER1L4) has been reported to be closely associated with the progression of various human cancers. However, the role of FER1L4 in OSCC remains unclear. METHODS: The expression level of FER1L4 in OSCC tissues and cancer cell lines was detected by using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was evaluated by cell counting kit-8 (CCK-8) assay and EdU staining assay. Cell invasion and migration were evaluated by Transwell assay. Cell apoptosis was detected by flow cytometry. Luciferase reporter assay was performed to determine the targeting relationship between FER1L4, miR-133a-5p and Prx1. The protein expression of Prx1 was detected by Western blot. In addition, a xenograft tumor model in vivo was constructed to confirm the function of FER1L4. RESULTS: FERIL4 was significantly upregulated in OSCC tissues and cancer cell lines. Moreover, high level of FER1L4 predicted a poor prognosis of OSCC patients. Silencing of FER1L4 not only significantly inhibited cell growth, invasion, migration and induced apoptosis in SCC-9 and HN4 cells in vitro, but also effectively suppressed the tumorigenesis of OSCC cells in vivo. Knockdown of FER1L4 significantly enhanced the expression of miR-133a-5p by sponging it, and then downregulated Prx1 expression. CONCLUSION: Our study elucidated a new mechanism of lncRNA FER1L4 that promoting OSCC progression by directly targeting miR-133a-5p/Prx1 axis and provided novel therapeutic targets for OSCC.

19.
Oncol Rep ; 45(1): 359-367, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33416121

RESUMO

Long non­coding RNA Fer­1­like protein 4 (FER1L4) has been reported to play crucial regulatory roles in tumor progression and apoptosis. However, its clinical significance and biological role in non­small cell lung cancer (NSCLC) are completely unknown. The purpose of this study was to investigate the expression of lncRNA FER1L4 in plasma and tissues of patients with NSCLC and study the mechanism of proliferation and apoptosis of lung cancer cells. The expression levels of FER1L4 in plasma and tissues of NSCLC patients and cell lines were analyzed via RT­qPCR. The effects of FER1L4 on cell proliferation, migration and invasion were analyzed by CCK­8, wound healing and Transwell assays, respectively. The expression levels of related proteins were detected by western blot assay, while cell apoptosis was determined by Hoechst staining and flow cytometry. The results revealed that FER1L4 was significantly downregulated in NSCLC plasma and tissues and lung cancer cell lines compared to corresponding controls. Moreover, a significant decrease of cell proliferation, migration and invasion were observed in FER1L4­overexpressed cells. FER1L4 could promote phosphatase and tension homolog deleted on chromosome ten (PTEN) and p53 expression, inhibit AKT phosphorylation expression, thus increasing the proportion of apoptotic cells. The present study indicated that FER1L4 may inhibit cell proliferation and promote apoptosis of NSCLC cells via the PTEN/AKT/p53 pathway, which provides a better understanding of the pathogenesis of NSCLC and may provide a novel potential therapeutic target for clinical treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , PTEN Fosfo-Hidrolase/genética , RNA Longo não Codificante/metabolismo , Adolescente , Adulto , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/sangue , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pulmão/patologia , Pulmão/cirurgia , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação/genética , Pneumonectomia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/análise , RNA Longo não Codificante/genética , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Adulto Jovem
20.
Front Cell Dev Biol ; 9: 631181, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33604341

RESUMO

Orthodontic tooth movement is achieved by periodontal tissue remodeling triggered by mechanical force. It is essential to investigate the reaction of periodontal ligament stem cells (PDLSCs) for improving orthodontic therapeutic approaches. Autophagy is an endogenous defense mechanism to prevent mechanical damage of environmental change. Long non-coding RNAs (lncRNAs) are key regulators in gene regulation, but their roles are still largely uncharacterized in the reaction of PDLSCs during orthodontic tooth movement. In this study, we showed that autophagy was significantly induced in PDLSCs under compressive force, as revealed by the markers of autophagy, microtubule-associated protein light chain 3 (LC3) II/I and Beclin1, and the formation of autophagosomes. After the application of compressive force, lncRNA FER1L4 was strongly upregulated. Overexpression of FER1L4 increased the formation of autophagosome and autolysosomes in PDLSCs, while knockdown of FER1L4 reversed the autophagic activity induced by mechanical force. In mechanism, FER1L4 inhibited the phosphorylation of protein kinase B (AKT) and subsequently increased the nuclear translocation of forkhead box O3 (FOXO3) and thus mediated autophagic cascades under compressive strain. In mouse model, the expression of Lc3 as well as Fer1l4 was increased in the pressure side of periodontal ligament during tooth movement. These findings suggest a novel mechanism of autophagy regulation by lncRNA during periodontal tissue remodeling of orthodontic treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA