Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Bioenerg Biomembr ; 56(1): 73-85, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37999809

RESUMO

Circular RNA (circRNA) plays multiple roles in the development of esophageal cancer (EC). Herein, we investigate the function of circ_0001944 in EC progression and the related mechanism. Expression of circ_0001944, microRNA-338-5p (miR-338-5p), pyruvate dehydrogenase kinase 1 (PDK1), E-cadherin and N-cadherin was analyzed by quantitative real-time polymerase chain reaction, Western blotting or immunohistochemistry assay. Cell viability, proliferation, apoptosis, invasion and migration were investigated by cell counting kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell invasion and wound-healing assays, respectively. Glucose consumption was detected by Glucose Assay Kit. Lactate production was analyzed by Lactate Assay Kit. ATP/ADP ratio was determined by ADP/ATP ratio Assay Kit. The associations among circ_0001944, miR-338-5p and PDK1 were identified by dual-luciferase reporter and RNA pull-down assays. Xenograft mouse model assay was used to explore the role of circ_0001944 on tumor tumorigenesis in vivo. Circ_0001944 and PDK1 expression were significantly upregulated, while miR-338-5p was downregulated in EC tissues and cells in contrast with normal esophageal tissues and cells. Circ_0001944 knockdown inhibited EC cell proliferation, invasion, migration and glycolysis but induced apoptosis. Meanwhile, circ_0001944 depletion suppressed tumor tumorigenesis in vivo. Mechanistically, circ_0001944 bound to miR-338-5p, and miR-338-5p targeted PDK1. In addition, miR-338-5p inhibitors attenuated circ_0001944 depletion-induced effects in EC cells. The regulation of miR-338-5p on EC progression involved the downregulation of PDK1. Further, circ_0001944 controlled PDK1 expression through miR-338-5p. Circ_0001944 knockdown inhibited EC development and glycolysis by regulating the miR-338-5p/PDK1 pathway, providing a promising target for EC therapy.


Assuntos
Neoplasias Esofágicas , MicroRNAs , Humanos , Animais , Camundongos , Neoplasias Esofágicas/genética , Carcinogênese , Glicólise , Proliferação de Células , Modelos Animais de Doenças , Glucose , Lactatos , Trifosfato de Adenosina , MicroRNAs/genética
2.
Cell Mol Biol Lett ; 28(1): 13, 2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36803975

RESUMO

BACKGROUND: Esophageal squamous carcinoma (ESCC) is a common malignancy that originates in the digestive tract. Lymph node metastasis (LNM) is a complicated process, and tumor lymphangiogenesis has been reported to be associated with the spread of tumor cells to lymph nodes (LNs), including in ESCC. However, little is currently known about the mechanisms involved in lymphangiogenesis in ESCC tumors. According to previous literature, we know that hsa_circ_0026611 expresses at a high level in serum exosomes of patients with ESCC and shows a close association with LNM and poor prognosis. However, details on the functions of circ_0026611 in ESCC remain unclear. We aim to explore the effects of circ_0026611 in ESCC cell-derived exosomes on lymphangiogenesis and its potential molecular mechanism. METHODS: We firstly examined how circ_0026611 may express in ESCC cells and exosomes by quantitative reverse transcription real-time polymerase chain reaction (RT-qPCR). The potential effects circ_0026611 may exert on lymphangiogenesis in ESCC cell-derived exosomes were assessed afterward via mechanism experiments. RESULTS: circ_0026611 high expression pattern was confirmed in ESCC cells and exosomes. ESCC cell-derived exosomes promoted lymphangiogenesis by transferring circ_0026611. Besides, circ_0026611 interacted with N-α-acetyltransferase 10 (NAA10) to inhibit NAA10-mediated prospero homeobox 1 (PROX1) acetylation with subsequent ubiquitination and degradation. Furthermore, circ_0026611 was verified to promote lymphangiogenesis in a PROX1-mediated manner. CONCLUSIONS: Exosomal circ_0026611 inhibited PROX1 acetylation and ubiquitination to promote lymphangiogenesis in ESCC.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , MicroRNAs , RNA Circular , Humanos , Acetilação , Linhagem Celular Tumoral , Proliferação de Células , Células Endoteliais/metabolismo , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Linfangiogênese/genética , Metástase Linfática , MicroRNAs/metabolismo , Acetiltransferase N-Terminal A/metabolismo , Acetiltransferase N-Terminal E/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitinação , RNA Circular/genética
3.
Acta Biochim Biophys Sin (Shanghai) ; 55(11): 1797-1805, 2023 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-37766459

RESUMO

LincRNA-P21 is a tumor suppressor in esophageal squamous cell carcinoma (ESCC). Cell adhesion modules play vital roles in cell-cell and cell-extracellular matrix (ECM) interactions and malignant cancer progression. In this study, we investigate whether lincRNA-P21 exerts its functions by regulating the cell adhesion molecule cadherin 5 (CDH5) in ESCC. Moreover, the RNA binding protein (RBP) mediators of lincRNA-P21 and CDH5 are further examined. Cell viability, growth and migratory ability are assessed by calcein-AM/PI double staining, CCK-8, EdU, Transwell, and wound healing assays. The expression of collagen I and fibronectin is examined by immunofluorescence (IF). LincRNA-P21 and CDH5 are quantified by RT-qPCR and western blot analysis. Potential lincRNA-P21 targets are identified by RNA sequencing. RBPs that can interact with lincRNA-P21 and CDH5 are identified by RNA immunoprecipitation (RIP) assay. LincRNA-P21 knockdown increases cell viability, growth, cell migration, and collagen I and fibronectin expression in ESCC cells. LincRNA-P21 depletion induces the dysregulation of 316 genes, including CDH5, in TE-1 cells. CDH5 is identified as a downstream molecule of lincRNA-P21 given its close correlation with cell adhesion, ECM reconstruction, and cancer progression. LincRNA-P21 exerts its functions by negatively regulating CDH5 expression. YTH domain containing 1 (YTHDC1) mediates the regulatory effect of lincRNA-P21 on CDH5. LincRNA-P21 knockdown elevates cell viability and growth, promotes cell migration, and induces ECM reorganization by upregulating CDH5 via RBP YTHDC1 in ESCC.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , RNA Longo não Codificante , Humanos , Carcinoma de Células Escamosas do Esôfago/metabolismo , Neoplasias Esofágicas/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Linhagem Celular Tumoral , Colágeno/genética , Colágeno/metabolismo , Proliferação de Células , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
4.
Biol Res ; 54(1): 7, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33653412

RESUMO

BACKGROUND: Leaves of the natural plant lotus are used in traditional Chinese medicine and tea production. They are rich in flavonoids. METHODS: In this study, lotus leaf flavonoids (LLF) were applied to human lung cancer A549 cells and human small cell lung cancer cells H446 in vitro to verify the effect of LLF on apoptosis in these cells through the ROS/p38 MAPK pathway. RESULTS: LLF had no toxic effect on normal cells at concentrations up to 500 µg/mL, but could significantly inhibit the proliferation of A549 cells and H446 cells. Flow cytometry showed that LLF could induce growth in A549 cells. We also found that LLF could increase ROS and MDA levels, and decrease SOD activity in A549 cells. Furthermore, qRT-PCR and western blot analyses showed that LLF could upregulate the expression of p38 MAPK (p-p38 MAPK), caspase-3, caspase-9, cleaved caspase-3, cleaved caspase-9 and Bax and downregulate the expression of Cu/Zn SOD, CAT, Nrf2, NQO1, HO-1, and Bcl-2 in A549 cells. Results of HPLC showed that LLF mainly contain five active substances: kaempferitrin, hyperoside, astragalin, phloridzin, and quercetin. The apoptosis-inducing effect of LLF on A549 cells came from these naturally active compounds. CONCLUSIONS: We have shown in this study that LLF is a bioactive substance that can induce apoptosis in A549 cells in vitro, and merits further research and development.


Assuntos
Apoptose/efeitos dos fármacos , Flavonoides/farmacologia , Lotus/química , Neoplasias Pulmonares/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células A549 , Proliferação de Células , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Compostos Fitoquímicos/farmacologia , Folhas de Planta/química , Transdução de Sinais/efeitos dos fármacos
6.
Tumour Biol ; 37(4): 4305-12, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26493997

RESUMO

The aim of this study is to investigate whether metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1) can be used as a potential therapy target for human esophageal squamous cell carcinoma. MALAT-1 expression levels were detected in 137 paired EC samples and adjacent nonneoplastic tissues. Human esophageal carcinoma cell lines EC9706 and KYSE150 were transfected with MALAT-1 small interference RNA. Cell proliferation, migration/invasion ability, cell cycle, and apoptosis were assessed. MALAT-1 expressed higher levels in esophageal cancer tissues when compared with paired adjacent normal tissues. This high expression was associated with a decreased survival rate. MALAT-1 knockdown induced a decrease in proliferation-enhanced apoptosis, inhibited migration/invasion, and reduced colony formation and led to cell cycle arrest at the G2/M phase. These data indicates that MALAT-1 could be exploited for therapeutic benefit.


Assuntos
Carcinoma de Células Escamosas/genética , Pontos de Checagem do Ciclo Celular/genética , Proliferação de Células/genética , Neoplasias Esofágicas/genética , RNA Longo não Codificante/biossíntese , Adulto , Idoso , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , RNA Longo não Codificante/genética
7.
Tumour Biol ; 35(5): 4697-704, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24443257

RESUMO

We conducted a perspective study to investigate whether the expression of excision repair cross-complementing 1 (ERCC1), xeroderma pigmentosum group G (XPG), breast cancer 1 (BRCA1), and ribonucleotide reductase M1 (RRM1) is correlated with clinical outcome of non-small cell lung cancer (NSCLC). Patients with histologically confirmed inoperable stages IIIB and IV NSCLC were collected and followed up until January 2012. Relative cDNA quantification for ERCC1, XPG, BRCA1, and RRM1 was performed using a fluorescence-based, real-time detection method. Cox regression analysis indicated that a high level of ERCC1 was associated with shorter overall survival (OS) and progression-free survival (PFS) times when compared with low expression, with adjusted hazard ratios (HRs) (95% confidence interval (CI)) of 2.25 (1.18-4.39) and 2.63 (1.33-5.25), respectively. High expression of BRCA1 was correlated with shorter OS and PFS times when compared with low expression, and the adjusted HRs (95% CI) were 3.29 (1.72-6.39) and 5.94 (2.80-13.06), respectively. Moreover, we found a significant correlation between BRCA1 expression and age (χ(2) = 4.14, P = 0.04) and stage (χ(2) = 5.26, P = 0.02). Our results suggest that ERCC1 and BRCA1 mRNA expressions are associated with PFS and OS in advanced NSCLC patients treated with platinum-based chemotherapy.


Assuntos
Proteína BRCA1/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Neoplasias Pulmonares/tratamento farmacológico , Adulto , Idoso , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Modelos de Riscos Proporcionais , RNA Mensageiro/análise , Fatores de Transcrição/genética , Resultado do Tratamento
8.
J Imaging Inform Med ; 37(4): 1529-1547, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38438696

RESUMO

Medical image segmentation is an important step in medical image analysis, especially as a crucial prerequisite for efficient disease diagnosis and treatment. The use of deep learning for image segmentation has become a prevalent trend. The widely adopted approach currently is U-Net and its variants. Moreover, with the remarkable success of pre-trained models in natural language processing tasks, transformer-based models like TransUNet have achieved desirable performance on multiple medical image segmentation datasets. Recently, the Segment Anything Model (SAM) and its variants have also been attempted for medical image segmentation. In this paper, we conduct a survey of the most representative seven medical image segmentation models in recent years. We theoretically analyze the characteristics of these models and quantitatively evaluate their performance on Tuberculosis Chest X-rays, Ovarian Tumors, and Liver Segmentation datasets. Finally, we discuss the main challenges and future trends in medical image segmentation. Our work can assist researchers in the related field to quickly establish medical segmentation models tailored to specific regions.


Assuntos
Redes Neurais de Computação , Humanos , Processamento de Imagem Assistida por Computador/métodos , Aprendizado Profundo , Feminino , Diagnóstico por Imagem/métodos , Neoplasias Ovarianas/diagnóstico por imagem , Fígado/diagnóstico por imagem
9.
Int J Biol Macromol ; 277(Pt 1): 133814, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38996888

RESUMO

The incidence of esophageal cancer continues to increase worldwide. Current therapeutic approaches have limited efficacy, so in order to search for better markers of the disease, it is necessary to further elucidate its molecular pathogenesis. Regulation of gene expression by long non-coding Rnas plays a role in many diseases, however the role in esophageal cancer is unclear. The aim of this study was to elucidate the role and regulatory mechanism of long non-coding RNA NRSN2-AS1 in the progression of esophageal cancer. By real-time quantitative PCR, immunohistochemistry, RNA interference, western blotting, and double luciferase reporter gene analysis, we found that NRSN2-AS1 was up-regulated in esophageal cancer tissues and cell lines, and was closely related to disease stage and prognosis. Functional studies have shown that the silencing of NRSN2-AS1 inhibits the proliferation of esophageal cancer cells, induces apoptosis, and prevents cell migration and invasion. In mouse models, NRSN2-AS1 also promoted tumor growth. The transcription factor TCFL5 upregulates the transcription of NRSN2-AS1, which acts as a sponge for microRNA(miR)-874-5p, thereby upregulating the expression of the oncogene RELT. Activation of the NRSN2-AS1/miR-874-5p/RELT regulatory axis was validated in vivo.


Assuntos
Proliferação de Células , Progressão da Doença , Neoplasias Esofágicas , Regulação Neoplásica da Expressão Gênica , MicroRNAs , RNA Longo não Codificante , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Camundongos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Masculino , Feminino , Apoptose/genética , Movimento Celular/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pessoa de Meia-Idade
10.
PLoS One ; 19(6): e0291531, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38924029

RESUMO

Tangeretin (Tan), a citrus flavonoid, possesses a strong anti-tumor efficacy in various human cancers. However, the precise role of Tan in the development of esophageal squamous cell carcinoma (ESCC) remains unclear. RNA sequencing (RNA-seq) analysis was performed to observe the Tan-related genes in Tan-treated TE-1 cells. The direct relationship between GLI family zinc finger 2 (GLI2) and the promoter of glycoprotein non-metastatic melanoma protein B (GPNMB) was predicted by bioinformatics analysis and validated by luciferase reporter and chromatin immunoprecipitation (ChIP) assays. Cell survival after Tan treatment was assessed by CCK8 assay. Gene expression levels were evaluated by a qRT-PCR, western blot, or immunofluorescence method. Cell migration and invasion were detected by wound-healing and transwell assays. The function of Tan in vivo was examined using xenograft studies. Our data indicated anti-migration and anti-invasion functions of Tan in ESCC cells in vitro. Tan also diminished tumor growth in vivo. Mechanistically, Tan diminished the expression and transcriptional activity of GLI2 in ESCC cells. Silencing of GLI2 resulted in decreased expression of GPNMB by inhibiting GPNMB transcription via the binding site at the GPNMB promoter at position +(1539-1550). Moreover, Tan down-regulated GPNMB expression in ESCC cells, and re-expression of GPNMB reversed anti-migration and anti-invasion functions of Tan in ESCC cells. Our findings uncover anti-migration and anti-invasion effects of Tan in ESCC cells by down-regulating GPNMB by suppressing GLI2-mediated GPNMB transcription, providing new evidence that Tan can function as a therapeutic agent against ESCC.


Assuntos
Movimento Celular , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Flavonas , Regulação Neoplásica da Expressão Gênica , Glicoproteínas de Membrana , Proteína Gli2 com Dedos de Zinco , Humanos , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Carcinoma de Células Escamosas do Esôfago/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Animais , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Linhagem Celular Tumoral , Camundongos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Flavonas/farmacologia , Movimento Celular/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Nus , Transcrição Gênica/efeitos dos fármacos , Regiões Promotoras Genéticas , Proliferação de Células/efeitos dos fármacos , Proteínas do Olho/metabolismo , Proteínas do Olho/genética , Proteínas Nucleares
11.
Cell Death Discov ; 9(1): 403, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37903782

RESUMO

Esophageal carcinoma (EC), one of the most lethal human malignancies, lacks effective targeted therapies. Indoleamine 2,3-dioxygenase 1 (IDO1) plays a key role in a variety of cancers, but its role and mechanism in EC are still unclear. Immunohistochemistry and qRT-PCR were used to analyze the expression of IDO1 in EC, and the prognostic value of IDO1 in EC was evaluated by Kaplan-Meier test. The in vitro and in vivo function loss/acquisition tests were performed to evaluate the biological effects of IDO1 in EC. The mechanism of action of IDO1-regulation EC was explored through Firefly luciferase & Renilla luciferase activity reporter, chromatin immunoprecipitation (ChIP) and immunofluorescence (IF) assays. Clinically, IDO1 expression was abnormally elevated in EC and positively correlated with overall survival. Functionally, IDO1 was contributed to the proliferation and migration of EC cells. Mechanically, IDO1 regulated the expression of chemokine C-X-C ligand 10 (CXCL10) by promoting the entry of NF-κB into the nucleus to combine with the promoter of CXCL10. Consistently, IDO1 facilitated EC progression may dependent on the presence of CXCL10. Moreover, NF-κB alleviated the inhibitory effect of IDO1 knockdown on EC. IDO1 drove the progression of EC by directly binding NF-κB and CXCL10, the finding that may provide an effective theoretical basis for precise therapies for EC.

12.
Thorac Cancer ; 13(9): 1299-1310, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35411716

RESUMO

BACKGROUND: Circular RNAs (circRNAs) can function as competing endogenous RNAs (ceRNAs) to impact the development of esophageal squamous cell cancer (ESCC). Human circ_0001946 has been identified as a potential anticancer factor in ESCC, yet our understanding of its molecular basis remains limited. METHODS: Circ_0001946, microRNA (miR)-1290 and SRY-box transcription factor 6 (SOX6) were quantified by quantitative reasl-time PCR (qRT-PCR) or immunoblotting. Cell proliferation was assessed by CCK-8 and EDU assays. Cell apoptosis and invasion were evaluated by flow cytometry and transwell assays, respectively. Cell migration was detected by transwell and wound-healing assays. The direct relationship between miR-1290 and circ_0001946 or SOX6 was determined by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Xenograft model assays were used to assess the role of circ_0001946 in tumor growth. RESULTS: Circ_0001946 expression was attenuated in human ESCC, and circ_0001946 increase impeded cell proliferation, invasion, migration and enhanced apoptosis in vitro. Moreover, circ_0001946 increase diminished xenograft growth in vivo. Mechanistically, circ_0001946 bound to miR-1290, and re-expression of miR-1290 reversed circ_0001946-dependent cell properties. SOX6 was a miR-1290 target and it was responsible for the regulation of miR-1290 in cell properties. Furthermore, circ_0001946 functioned as a ceRNA to regulate SOX6 expression via miR-1290. CONCLUSION: Our findings uncover an undescribed molecular mechanism, the circ_0001946/miR-1290/SOX6 ceRNA crosstalk, for the anti-ESCC activity of circ_0001946.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , MicroRNAs , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Células Epiteliais/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Fatores de Transcrição SOXD/genética , Fatores de Transcrição SOXD/metabolismo
13.
J Gastrointest Oncol ; 13(2): 499-509, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35557578

RESUMO

Background: Lower frequency of tooth brushing was thought to be associated with esophageal carcinoma (EC). However, some researchers suggested that this association did not exist or had not yet reached statistical significance. The purpose of this study was to calculate a more precise estimation of the relationship between the frequency of tooth brushing and the risk of EC by combining the results between different studies using the meta-analysis. Methods: We searched the PubMed, Embase, Web of Science, and Scopus electronic databases up to July 2021. According to PECO approach (Population, Exposure, Comparator and Outcomes), we assessed the association between tooth brushing frequency and EC risk which reported the adjusted risk ratios (adjRR), hazard ratios (adjHR), or odds ratios (adjOR) with 95% confidence interval (CI). The random effects model was used to quantitatively evaluate the combined results. Two researchers independently evaluated the risk bias of the included studies using the Newcastle-Ottawa Scale (NOS). The robustness of results was evaluated by subgroup analysis, sensitivity analysis, and publication bias. Results: In total, we identified 13 articles with 14 case-control studies which included 16,773 participants and 5,673 patients. Pooled results showed the lowest frequency of brushing was significantly associated with an increased risk of EC in comparison to the highest (adjOR: 2.00, 95% CI: 1.61-2.48). There was moderate heterogeneity among included studies (P=0.001, I2=61.4%). The original studies included in this meta-analysis were all case-control studies. Study quality was all moderate or above based on NOS score ranges of 6 stars or more. Conclusions: Available evidence suggests a low frequency of tooth brushing may be an important risk factor for EC. However, higher quality studies should continue to be conducted to investigate the optimal threshold of brushing frequency for the prevention of EC.

14.
Cell Cycle ; 20(12): 1163-1172, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34057012

RESUMO

This study aimed to explore the role of micorRNA-2053 in esophageal cancer development. The expression level of miR-2053 in esophageal cancer cell lines was detected. After cell transfection, the effects of miR-2053 overexpression on proliferation, apoptosis, migration and invasion of esophageal cancer cells were determined. Moreover, the potential molecular mechanism was explored by measuring the epithelial-mesenchymal transition (EMT) and apoptosis-related proteins. Luciferase reporter assay was conducted to investigate the target gene of miR-2053. The protein expressions of PI3K/AKT pathway associated factors were detected after overexpression of miR-2053 or administration with the pathway inhibitor LY294002. The miR-2053 was downregulated in esophageal cancer cell lines. Overexpression of miR-2053 inhibited cell proliferation, migration and invasion while promoted apoptosis. Molecular mechanism elucidated that miR-2053 could reduce EMT and elevate the expression of pro-apoptotic proteins. Further study found that overexpressed miR-2053 could negatively regulate KIF3C and involve in PI3K/AKT signaling pathway. Our study demonstrated the downregulation of miR-2053 in esophageal cancer. Downregulation of miR-2053 involved in the proliferation, apoptosis, migration and invasion of esophageal cancer cells through upregulating KIF3C expression and activating the PI3K/AKT signaling pathway. miR-2053 may have the potential in clinical treatment of esophageal cancer.


Assuntos
Carcinogênese/metabolismo , Progressão da Doença , Neoplasias Esofágicas/metabolismo , Cinesinas/metabolismo , MicroRNAs/metabolismo , Transdução de Sinais/genética , Apoptose/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Cromonas/farmacologia , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Humanos , MicroRNAs/genética , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transfecção , Regulação para Cima/genética
15.
Int J Mol Med ; 48(1)2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34080641

RESUMO

Excessive inflammatory response and apoptosis play key roles in the pathogenic mechanisms of sepsis­induced acute lung injury (ALI); however, the molecular pathways linked to ALI pathogenesis remain unclear. Recently, microRNAs (miRNAs/miRs) have emerged as important regulators of inflammation and apoptosis in sepsis­induced ALI; however, the exact regulatory mechanisms of miRNAs remain poorly understood. In the present study, the gene microarray dataset GSE133733 obtained from the Gene Expression Omnibus database was analyzed and a total of 38 differentially regulated miRNAs were identified, including 17 upregulated miRNAs and 21 downregulated miRNAs, in mice with lipopolysaccharide (LPS)­induced ALI, in comparison to the normal control mice. miR­129 was found to be the most significant miRNA, among the identified miRNAs. The upregulation of miR­129 markedly alleviated LPS­induced lung injury, as indicated by the decrease in lung permeability in and the wet­to­dry lung weight ratio, as well as the improved survival rate of mice with ALI administered miR­129 mimic. Moreover, the upregulation of miR­129 reduced pulmonary inflammation and apoptosis in mice with ALI. Of note, transforming growth factor activated kinase­1 (TAK1), a well­known regulator of the nuclear factor­κB (NF­κB) pathway, was directly targeted by miR­129 in RAW 264.7 cells. More importantly, miR­129 upregulation impeded the LPS­induced activation of the TAK1/NF­κB signaling pathway, as illustrated by the suppression of the nuclear phosphorylated­p65, p­IκB­α and p­IKKß expression levels. Collectively, the findings of the present study indicate that miR­129 protects mice against sepsis­induced ALI by suppressing pulmonary inflammation and apoptosis through the regulation of the TAK1/NF­κB signaling pathway. This introduces the basis for future research concerning the application of miR­129 and its targets for the treatment of ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , MAP Quinase Quinase Quinases/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Sepse/metabolismo , Transdução de Sinais , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/prevenção & controle , Animais , Bases de Dados de Ácidos Nucleicos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/prevenção & controle , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Sepse/induzido quimicamente , Sepse/prevenção & controle
16.
Cell Cycle ; 20(13): 1231-1241, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34121605

RESUMO

The members of the tumor necrosis factor receptor (TNFR) family have been demonstrated to play critical roles in various cancers. However, little is known about the function of the Receptor Expressed in Lymphoid Tissues (RELT) in cancers, which is a member of the TNFR family, especially in the esophageal squamous cell carcinoma (ESCC). In this study, we found that RELT expression was increased in ESCC tissues and was consequently associated with poor overall survival of ESCC patients. Moreover, RELT overexpression was found to promote cell growth, cell cycle progression, and suppressed cell apoptosis in vitro; it also decreased the expression of p27 and caspase 3, and increased the expression of survivin. In addition, RELT contributed to the tumorigenesis of ESCC in vivo. Furthermore, we suggest that RELT may function in the pathogenesis of ESCC by activating the nuclear factor κB (NF-κB) pathway. An inhibitor of NF-κB reversed the RELT-induced malignancy in the ESCC cells. Altogether, our findings identified that RELT served as an oncogene in ESCC through the NF-κB pathway, suggesting that RELT may be developed as a novel biomarker for the diagnosis and treatment of the ESCC.


Assuntos
Proliferação de Células , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/genética , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Survivina/metabolismo , Carga Tumoral
17.
Front Oncol ; 11: 761346, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34820329

RESUMO

BACKGROUND: Increasing researches have been reported that epigenetic alterations play critical roles in ESCC development. However, the role of the histone demethylase KDM4D in ESCC tumorigenesis is poorly investigated. This study aims to discover the underlying mechanisms between KDM4D and ESCC progression. METHODS: CCK-8 assays, clone formation assay and soft-agar assays were performed to assess cell proliferation. Transwell assay was utilized to assess cell migration efficiency, while sphere formation assay was used to evaluate the cell self-renewal ability. Bioinformatic analysis was conducted to identify prognostic factors and predict the potential E3 ubiquitin ligases. In vitro ubiquitination assay was conducted to confirm the regulations between SYVN1 and HMGB1. The mRNA levels or protein levels of genes were detected by real-time PCR and western blot analysis. In vivo tumor xenograft models were used to determine whether the HMGB1 inhibition affected the malignant features of ESCC cells. RESULT: Epigenome screening and low-throughput validations highlighted that KDM4D is a tumor suppressor in ESCC. KDM4D expressed lowly in tumors that predicts poor prognosis. KDM4D deficiency significantly enhanced tumor growth, migration and stemness. Mechanistically, KDM4D transcriptionally activates SYVN1 expressions via H3K9me3 demethylation at the promoter region, thereby triggering the ubiquitin-dependent degradation of HMGB1. Low KDM4D depended on accumulated HMGB1 to drive ESCC progression and aggressiveness. Targeting HMGB1 (Glycyrrhizin) could remarkably suppress ESCC tumor growth in vitro and in vivo, especially in KDM4D-deficient cells. CONCLUSIONS: We systematically identified KDM4D/SYVN1/HMGB1 axis in ESCC progression, proving novel biomarkers and potential therapeutic targets.

18.
Front Oncol ; 11: 780938, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34966683

RESUMO

BACKGROUND: CircPVT1 is demonstrated to promote cancer progression in esophageal squamous cell carcinoma (ESCC). However, the role and potential functional mechanisms of circPVT1 in regulating 5-fluorouracil (5-FU) chemosensitivity remain largely unknown. METHODS: ESCC cells resistant to 5-FU were induced with continuous increasing concentrations of 5-FU step-wisely. A cell counting kit-8 assay was used to analyze the viability of ESCC cells. LDH release assay kit was used to evaluate the cytotoxicity. RT-qPCR was used to assess the expression level of non-coding RNAs and cDNAs. Luciferase was used to confirm the interaction between non-coding RNAs and targets. Western blotting was used to detect the expression of downstream signaling proteins. Flow cytometry and ferroptosis detection assay kit were utilized to measure the ferroptosis of ESCC cells. RESULTS: CircPVT1 was significantly upregulated in ESCC cells resistant to 5-FU. Knockdown of circPVT1 enhanced the 5-FU chemosensitivity of ESCC cells resistant to 5-FU by increasing cytotoxicity and downregulating multidrug-resistant associated proteins, including P-gp and MRP1. Luciferase assay showed that circPVT1 acted as a sponge of miR-30a-5p, and Frizzled3 (FZD3) was a downstream target of miR-30a-5p. The enhanced 5-FU chemosensitivity by circPVT1 knockdown was reversed with miR-30a-5p inhibitor. Besides, the increased 5-FU chemosensitivity by miR-30a-5p mimics was reversed with FZD3 overexpression. Furthermore, knockdown of circPVT1 increased ferroptosis through downregulating p-ß-catenin, GPX4, and SLC7A11 while miR-30a-5p inhibition and FZD3 overexpression reversed the phenotype by upregulating p-ß-catenin, GPX4, and SLC7A11. CONCLUSIONS: These results suggested a key role for circPVT1 in ESCC 5-FU-chemosensitivity in regulating the Wnt/ß-catenin pathway and ferroptosis via miR-30a-5p/FZD3 axis, which might be a potential target in ESCC therapy.

19.
Onco Targets Ther ; 13: 6987-6996, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32764989

RESUMO

BACKGROUND: The aim of this study was to observe the preventive effect of flavonoids extracted from Malus asiatica Nakai leaves (FMANL) on esophageal cancer in mice, especially the ability of FMANL to regulate the interleukin 17 (IL-17) signaling pathway during this process. MATERIALS AND METHODS: The C57BL/6J mice were treated with 4-nitroquinoline N-oxide (4NQO) to induce esophageal cancer, and the visceral tissue index and the serum and esophageal tissue indexes of mice were used to verify the effect of FMANL. RESULTS: The experimental results showed that FMANL can effectively control the changes in visceral tissue caused by esophageal cancer. FMANL could increase the cytokine levels of interleukin 10 (IL-10), monocyte chemotactic protein 1 (MCP-1) and decrease the cytokine levels of tumor necrosis factor alpha (TNF-α), interferon-γ (IFN-γ), interleukin 6 (IL-6), and interleukin 12p70 (IL-12p70) in serum of mice with esophageal cancer. FMANL could also reduce CD3+, CD4+, and CD8+ and enhance CD19+ mouse peripheral blood lymphocytes. The results of qPCR and Western blot analysis showed that FMANL could down-regulate the mRNA and protein expression levels of IL-17, interleukin 23 (IL-23), interleukin 1 beta (IL-1ß), chemokine (C-X-C) ligand 1 (CXCL1), chemokine (C-X-C) ligand 2 (CXCL2), S100 calcium-binding protein A8 (S100A8), S100 calcium-binding protein A9 (S100A9), matrix metalloprotein 9 (MMP-9), and matrix metalloprotein 13 (MMP-1) in mice with esophageal cancer. High-performance liquid chromatography (HPLC) detection showed that FMANL contained 10 chemicals, including rutin, hyperoside, isoquercitrin, dihydroquercetin, quercitrin, hesperidin, myricetin, baicalin, neohesperidin dihydrochalcone, and quercetin. CONCLUSION: It could be concluded that FMANL can effectively prevent experimentally induced esophageal cancer in mice, and its effects might be obtained from 10 compounds present in FMANL.

20.
Mol Med Rep ; 20(4): 3633-3641, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31485658

RESUMO

As a tumor­associated microRNA (miR), miR­212 has dual functions; either as an oncogene or a tumor suppressor. A high expression level of miR­212 was reported to be associated with poor outcome in patients with esophageal squamous cell carcinoma (ESCC), however, its role in ESCC progression has not been explored. In the present study, an in vitro cell model of lentivirus­mediated gain­of­function demonstrated promotion of ESCC cell migration and invasion when miR­212 was overexpressed, and no effect on cell proliferation. miR­212 resulted in downregulation of the expression of E­cadherin, ß­catenin, vimentin and Twist1. Moreover, it led to increased levels of extracellular matrix (ECM)­degrading enzymes, matrix metalloproteinase­9 and urokinase­type plasminogen activator. Furthermore, berberine inhibited miR­212­induced ESCC cell migration, unlike the PI3K inhibitor LY294002, rapamycin (mTOR inhibitor), 5­(Tetradecyloxy)­2­furoic acid (TOFA; an acetyl­CoA carboxylase 1 inhibitor), metformin and propranolol. These data suggest that miR­212 activates multiple signaling cascades and facilitates ESCC cell motility and invasion by promoting the epithelial­mesenchymal transition and degrading the ECM. Berberine may be a potential therapeutic agent against metastasis in patients with ESCC, who express high levels of miR­212.


Assuntos
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Invasividade Neoplásica/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Humanos , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA