Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38687856

RESUMO

Objective: Given the high incidence of pulmonary complications and poor prognosis in patients with multiple rib fractures, we have developed a risk prediction model for pulmonary complications in patients with MRF. In order to identify the high-risk groups prone to pulmonary complications as early as possible, we will intervene in advance and provide targeted interventions to improve patients' quality of life and disease prognosis. Methods: The prospective cohort study design scheme was used to collect data information based on the hospital's electronic medical record system. The constructed cohort included 314 cases, and the validation cohort included 119 patients with MRF. The risk prediction model for pulmonary complications in patients with MRF was established using the backward screening method and multivariate logistic regression analysis. The predictive quality and clinical utility of the model were assessed using AUC, sensitivity, specificity, calibration curves, and clinical decision curves. Results: Seven predictors were finally included after multivariate logistic regression analysis: age, smoking history, diabetes mellitus, presence of other fracture combinations, serum albumin, treatment modality, and presence of underlying lung disease. The construction of the cohort yielded an AUC of 0.928 (95% CI 0899-0.956; P < .001) for the present model, with a sensitivity of 81.2% and a specificity of 76.8%, while the external validation cohort yielded an AUC of 0.942 (95% CI 0.900-0.983; P < .001), with a sensitivity of 76.7% and a specificity of 78.4%, and the H-L chi-squared tests all showed P > .05. Conclusions: The column-line diagram model of pulmonary complications in patients with MRF constructed in this study showed good discriminative and calibration performance in both internal and external validation, which is helpful for clinicians to identify individuals at high risk of pulmonary complications as early as possible, and thus can be recommended for clinical use.

2.
Clin Exp Pharmacol Physiol ; 47(3): 503-516, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31788833

RESUMO

Colorectal cancer (CRC), a leading cause of cancer death, has recently been known as the most prevalent malignancy worldwide. Although chemotherapy is an important therapeutic option for CRC patients, multidrug resistance (MDR) still remains a major cause of chemotherapy failure. Transmembrane protein 45A (TMEM45A) has been found highly expressed in various cancers, and is also proposed as an interesting biomarker for chemoresistance. However, the association between TMEM45A and MDR in CRC remains unclear. This study aimed to investigate the key role of TMEM45A in CRC by knockdown of its expression in 5-FU-resistant CRC cells (HCT-8/5-FU and SW480/5-FU) and their parental cells (HCT-8 and SW480). Data showed that TMEM45A was significantly up-regulated in HCT-8/5-FU and SW480/5-FU cells in comparison with their parental HCT-8 and SW480 cells. Knockdown of TMEM45A enhanced 5-FU sensitivity and 5-FU-induced apoptosis in HCT-8/5-FU and SW480/5-FU cells. It was also found that inhibition of TMEM45A increased the intracellular accumulation of Rhodamine-123 and down-regulated the expression of MDR1 in HCT-8/5-FU and SW480/5-FU cells. In addition, knockdown of TMEM45A suppressed migration and invasion of HCT-8/5-FU and SW480/5-FU cells. Furthermore, knockdown of TMEM45A not only attenuated MDR-enhanced epithelial-mesenchymal transition (EMT), but also suppressed MDR-enhanced activation of the TGF-ß signalling pathway in HCT-8/5-FU and SW480/5-FU cells. Taken together, our study suggests that knockdown of TMEM45A can effectively overcome MDR and inhibit EMT via suppression of the TGF-ß signalling pathway in human CRC cells, and that targeting TMEM45A will be a potential strategy in the treatment of MDR in CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Resistência a Múltiplos Medicamentos/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Proteínas de Membrana/deficiência , Fator de Crescimento Transformador beta/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Relação Dose-Resposta a Droga , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fluoruracila/farmacologia , Técnicas de Silenciamento de Genes/métodos , Humanos , Proteínas de Membrana/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores
3.
J Cell Physiol ; 232(7): 1879-1884, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27966796

RESUMO

Colorectal cancer is the second most deadly malignancy in the United States. However, the currently screening options had their limitation. Novel biomarkers for colorectal cancer detections are necessary to reduce the mortality. The clinical information, mRNA expression levels and DNA methylation information of colorectal cancer were downloaded from TCGA. The patients were separated into training group and testing group based on their platforms for DNA methylation. Beta values of DNA methylation from tumor tissues and normal tissues were utilized to figure out the position that were differentially methylated. The expression levels of mRNA of thirteen genes, whose CpG islands were differentially methylated, were extracted from the RNA-Seq results from TCGA. The probabilities whether the mRNA was differentially expressed between tumor and normal samples were calculated using Student's t-test. Logistic regression and decision tree were built for cancer detection and their performances were evaluated by the area under the curve (AUC). Twenty-four genomic locations were differentially methylated, which could be mapped to eleven genes. Nine out of eleven genes had differentially expressed mRNA levels, which were used to build the model for cancer detection. The final detection models consisting of mRNA expression levels of these nine genes had great performances on both training group and testing group. The model that constructed in this study suggested MSX1 and DCLK1 might be used in colorectal cancer detection or as target of cancer therapies. J. Cell. Physiol. 232: 1879-1884, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Metilação de DNA/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fator de Transcrição MSX1/genética , Proteínas Serina-Treonina Quinases/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG/genética , Árvores de Decisões , Quinases Semelhantes a Duplacortina , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Logísticos , Fator de Transcrição MSX1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Curva ROC
4.
Front Surg ; 8: 790380, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35071313

RESUMO

Objective: It has been shown that LIM-domain-binding protein 1 (LDB1) is involved in the tumorigenesis of several cancers, but its function in colorectal cancer (CRC) has not been fully explained. This study is aimed to investigate whether LDB1 is involved in regulating the cell growth and drug sensitivity of CRC. Methods: To analyze the protein expression of LDB1 in CRC tissues, western blot was used. KM plotter and UALCAN databases were used to predict the prognosis of CRC patients with low or high LDB1 expression. To do the correlation analysis in CRC tissues, GEPIA database was used. CCK-8 assay and xenograft models were used to evaluate the effects of LDB1 in CRC cell growth. An oxaliplatin-resistant cell line was constructed to evaluate the effect of LDB1 in drug sensitivity of CRC cells. Results: Our current research confirmed that LDB1 was upregulated in CRC tumor tissues, and its elevation predicted a poor prognosis for CRC patients. LDB1 was also found positively correlated with CCNA1, BCL2 and BCLW, but negatively correlated with the pro-apoptotic signals (BID, BAX and BAK). Silence of LDB1 significantly inhibited CRC cell growth in vitro, and CRC cells with low expression of LDB1 had a lower tumorigenesis rate in tumor-bearing nude mice. Our experiments also showed that LDB1 silence enhanced the anti-tumor activity of oxaliplatin in CRC cells. The expression of LDB1 was also found increased in oxaliplatin-resistant CRC cell lines, and silence of LDB1 partly restored the antitumor effect of oxaliplatin in an oxaliplatin-resistant CRC cell line. Conclusion: Our current results revealed the roles of LDB1 in the growth and drug resistance of CRC cells, and may provide the new theoretical support for LDB1 as a potential target for the treatment of CRC in the future.

5.
Onco Targets Ther ; 13: 371-379, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32021282

RESUMO

BACKGROUND AND OBJECTIVES: RING finger protein 38 (RNF38) has been reported to be involved in the tumorigenesis of several tumors, but its role in colorectal cancer (CRC) is still not investigated. In the present study, we aimed to investigate the effect of RNF38 in CRC cells. MATERIALS AND METHODS: The public tumor databases GEPIA and Kaplan-Meier Plotter were used to analyze RNF38 expression and patients' overall survival in CRC. The qRT-PCR was carried out to assess the mRNA levels of RNF38 and LDB1. Western blot and co-immunoprecipitation were used to detect protein expression and ubiquitination. CCK-8 assay was performed to analyze CRC cell growth and viability. RESULTS: RNF38 was found downregulated in CRC tumor tissues and cell lines, and CRC patients with high RNF38 expression had a longer overall survival than patients with low RNF38 expression. Our further investigations showed that RNF38 interacted with LDB1, and downregulated LDB1 expression by inducing its polyubiquitination. Moreover, overexpression of RNF38 inhibited CRC cell growth but enforced LDB1 could significantly antagonize RNF38-induced cell growth inhibition in CRC cells. Additionally, RNF38/LDB1 axis was involved in the drug sensitivity of 5-FU to CRC cells. CONCLUSION: Our studies suggested that RNF38 was functional in CRC cells, and downregulated CRC cell growth by inducing LDB1 polyubiquitination, which indicated that RNF38 could be as a novel target for CRC therapy.

6.
J BUON ; 24(5): 1830-1836, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31786844

RESUMO

PURPOSE: To elucidate the potential role of long non-coding RNA (lncRNA) DUXAP8 in the malignant progression of colorectal cancer (CRC) and its possible molecular mechanism. METHODS: The expression level of lncRNA DUXAP8 in CRC tissues and matched paracancerous tissues was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Meanwhile, its level in CRC patients with different tumor sizes and tumor grades was determined. The regulatory effects of DUXAP8 on the behaviors of CRC cells were evaluated by cell counting kit-8 (CCK-8), 5-Ethynyl-2'- deoxyuridine (EdU) and Transwell assay. The interaction between LSD1, EZH2 and DUXAP8 was evaluated by RNA-protein interactions and RIP assay. Linear regression analyses were conducted to examine the correlation between DUXAP8 and LSD1, EZH2. RESULTS: LncRNA DUXAP8 was upregulated in CRC tissues and cell lines. Its level remained higher in CRC with larger tumor size or higher tumor grade. Knockdown of DUXAP8 suppressed the proliferative, migratory and invasive abilities of DLD-1 and SW480 cells. Both RF classifier and SVM classifier predicted the pronounced accuracies of LSD1 and EZH2. RIP assay further demonstrated the interaction between DUXAP8 and LSD1, EZH2. Knockdown of LSD1 or EZH2 could attenuate the proliferative rate of CRC cells. Moreover, the mRNA levels of LSD1 and EZH2 were positively correlated with DUXAP8 in CRC. CONCLUSIONS: LncRNA DUXAP8 accelerates the malignant progression of CRC via positively regulating EZH2 and LSD1.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Colorretais/enzimologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histona Desmetilases/metabolismo , RNA Longo não Codificante/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/genética , Humanos , Invasividade Neoplásica , RNA Longo não Codificante/genética , Transdução de Sinais , Fatores de Tempo
7.
Kaohsiung J Med Sci ; 35(8): 467-473, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31063232

RESUMO

MicroRNA-26a-5p (miR-26a-5p) has been reported to be involved in the tumorigenesis of several tumors, but its function in breast cancer is still unknown. In this study, miR-26a-5p was found significantly downregulated in both of the breast cancer tissues and cell lines, and low expression of miR-26a-5p predicted a poor prognosis for breast cancer patients. Overexpression of miR-26a-5p could significantly inhibit breast cancer cell growth. Further studies revealed that overexpression of miR-26a-5p downregulated the protein levels of Cyclin D1, CDK4, and CDK6, but upregulated the expression levels of p21, p27, and p53. In mechanism, miR-26a-5p targeted the 3'UTR of ring finger protein 6 (RNF6) mRNA and inhibited RNF6 expression in breast cancer cells. Moreover, overexpression of miR-26a-5p inhibited RNF6/ERα/Bcl-xL axis in breast cancer cells. In contrast, inhibiting miR-26a-5p upregulated RNF6/ERα/Bcl-xL axis. Further studies indicated that miR-26a-5p mediated RNF6/ERα/Bcl-xL axis through regulating the stability of ERα protein. Collectively, downregulation of miR-26a-5p plays essential roles in breast cancer by mediating RNF6/ERα/Bcl-xL axis, which might provide important implications for the therapeutics of breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas de Ligação a DNA/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , MicroRNAs/genética , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA