Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Mol Biotechnol ; 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38619801

RESUMO

N6-methyladenosine (m6A) is a common posttranscriptional RNA modification and plays an important role in cancer biology. Circular RNAs (circRNAs) are also reported to participate in lung adenocarcinoma (LUAD) progression. Here, we aimed to investigate the functions of Wilms tumor 1-associating protein (WTAP) methyltransferase and circEEF2 in LUAD cell tumorigenesis, and probe whether circEEF2 functioned through WTAP-induced m6A modification and its potential mechanisms. Functional analyses were conducted by tube formation, sphere formation, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and transwell assays in vitro as well as tumor formation experiments in mice, respectively. The N6-methyladenine (m6A) modification in circEEF2 mRNA was determined by RNA immunoprecipitation (Me-RIP) assay. The interaction between IGF2BP2 (Insulin Like Growth Factor 2 MRNA-Binding Protein 2) and circEEF2 or Calcium-activated nucleotidase 1 (CANT1) mRNA was confirmed using RIP assay. LUAD tissues and cells showed high circEEF2 expression, and the deficiency of circEEF2 suppressed LUAD cell angiogenesis, stemness, proliferation, migration, and invasion. WTAP induced circEEF2 m6A modification. WTAP silencing repressed the oncogenic phenotypes of LUAD cells via stabilizing circEEF2 in an m6A-dependent manner. IGF2BP2 interacted with circEEF2 and CANT1, and WTAP and circEEF2 could regulate CANT1 expression through IGF2BP2. The inhibition of LUAD cell oncogenic phenotypes caused by circEEF2 deficiency was abolished by CANT1 overexpression. In addition, WTAP silencing impeded LUAD growth via modulating circEEF2 and CANT1 in vivo. WTAP-mediated m6A modification of circEEF2 promotes lung adenocarcinoma growth and tumorigenesis by stabilizing CANT1 through IGF2BP2.

2.
J Microbiol Biotechnol ; 34(2): 249-261, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38419324

RESUMO

New anti-lung cancer therapies are urgently required to improve clinical outcomes. Since ganodermanontriol (GDNT) has been identified as a potential antineoplastic agent, its role in lung adenocarcinoma (LUAD) is investigated in this study. Concretely, lung cancer cells were treated with GDNT and/or mycophenolate mofetil (MMF), after which MTT assay, flow cytometry and Western blot were conducted. Following bioinformatics analysis, carboxylesterase 2 (CES2) was knocked down and rescue assays were carried out in vitro. Xenograft experiment was performed on mice, followed by drug administration, measurement of tumor growth and determination of CES2, IMPDH1 and IMPDH2 expressions. As a result, the viability of lung cancer cells was reduced by GDNT or MMF. GDNT enhanced the effects of MMF on suppressing viability, promoting apoptosis and inducing cell cycle arrest in lung cancer cells. GDNT up-regulated CES2 level, and strengthened the effects of MMF on down-regulating IMPDH1 and IMPDH2 levels in the cells. IMPDH1 and IMPDH2 were highly expressed in LUAD samples. CES2 was a potential target for GDNT. CES2 knockdown reversed the synergistic effect of GDNT and MMF against lung cancer in vitro. GDNT potentiated the role of MMF in inhibiting tumor growth and expressions of CES2 and IMPDH1/2 in lung cancer in vivo. Collectively, GDNT suppresses the progression of LUAD by activating CES2 to enhance the metabolism of MMF.


Assuntos
Adenocarcinoma de Pulmão , Antineoplásicos , Lanosterol/análogos & derivados , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Ácido Micofenólico/farmacologia , Antineoplásicos/farmacologia , Adenocarcinoma de Pulmão/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Carboxilesterase
3.
J Pharm Pharmacol ; 76(4): 354-367, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38330446

RESUMO

OBJECTIVES: Reportedly, ganoderic acid A (GA-A) increases the sensitivity of hepatocellular carcinoma cells to cisplatin (DDP) chemotherapy. Therefore, this study aims to fathom the influence of GA-A on lung cancer cells. METHODS: After the construction of A549/DDP cells through exposure to DDP, the effects of GA-A on A549 and A549/DDP cells were revealed by cellular functional assays, western blot and quantitative reverse transcription PCR (qRT-PCR). The DDP-resistant lung cancer tumor was established in vivo, followed by further validation of the mechanism of GA-A. RESULTS: GA-A suppressed the viability, migration, and invasion while downregulating Beclin and autophagy marker LC3II/LC3I levels and upregulating P62 levels in A549 and A549/DDP cells. These effects were reversed by circFLNA overexpression. Also, GA-A reinforced the sensitivity of A549/DDP cells to DDP, elevated the apoptosis and regulated the circFLNA/miR-486-3p/cytochrome P450 family 1 subfamily A member 1 (CYP1A1)/X-ray repair cross-complementing 1 (XRCC1) axis. The reversal effects of circFLNA overexpression on GA-A-induced viability and apoptosis of A549/DDP cells could all be counteracted in the presence of 3MA. GA-A inhibited lung cancer tumor growth and blocked autophagy. CONCLUSION: GA-A suppresses autophagy by regulating the circFLNA/miR-486-3p/CYP1A1/XRCC1 axis to strengthen the sensitivity of lung cancer cells to DDP.


Assuntos
Antineoplásicos , Autofagia , Carcinoma Pulmonar de Células não Pequenas , Ácidos Heptanoicos , Lanosterol , Neoplasias Pulmonares , MicroRNAs , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/farmacologia , Citocromo P-450 CYP1A1/efeitos dos fármacos , Citocromo P-450 CYP1A1/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ácidos Heptanoicos/farmacologia , Ácidos Heptanoicos/uso terapêutico , Lanosterol/análogos & derivados , Lanosterol/farmacologia , Lanosterol/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MicroRNAs/efeitos dos fármacos , MicroRNAs/metabolismo , RNA Circular/efeitos dos fármacos , RNA Circular/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/efeitos dos fármacos , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/metabolismo
4.
Growth Factors ; : 1-10, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38164009

RESUMO

Non-small cell lung cancer (NSCLC) stands prominent among the prevailing and formidable oncological entities. The immune and metabolic-related molecule Phospholipase A2, group IID (PLA2G2D) exerts promotional effects on tumor progression. However, its involvement in cancer angiogenesis remains elusive. Therefore, this investigation delved into the functional significance of PLA2G2D concerning angiogenesis in NSCLC. This study analyzed the expression and enriched pathways of PLA2G2D in NSCLC tissues through bioinformatics analysis, and measured the expression of PLA2G2D in NSCLC cells using qRT-PCR and western blot (WB). Subsequently, the viability and angiogenic potential of NSCLC cells were assessed employing CCK-8 and angiogenesis assays, respectively. The expression profile of angiogenic factors was analyzed through WB. Finally, the expression of glycolysis pathway-related genes, extracellular acidification rate and oxygen consumption rate, and the levels of pyruvate, lactate, citrate, and malate were analyzed in NSCLC cells using qRT-PCR, Seahorse XF 96, and related kits. Bioinformatics analysis revealed the upregulation of PLA2G2D in NSCLC tissues and its association with VEGF and glycolysis signaling pathways. Molecular and cellular experiments demonstrated that upregulated PLA2G2D promoted the viability, angiogenic ability, and glycolysis pathway of NSCLC cells. Rescue assays revealed that the effects of high expression of PLA2G2D on the viability, angiogenic ability, and glycolysis of NSCLC cells were weakened after the addition of the glycolysis inhibitor 2-DG. In summary, PLA2G2D plays a key role in NSCLC angiogenesis through aerobic glycolysis, displaying great potential as a target for anti-angiogenesis therapy.

5.
Artigo em Inglês | MEDLINE | ID: mdl-38265409

RESUMO

BACKGROUND: Lung cancer (LC) is primarily responsible for cancer-related deaths worldwide. Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells acquire mesenchymal features and is associated with the development of tumors. CBX8, a member of the PcG protein family, plays a critical role in various cancers, containing LC. However, specific regulatory mechanisms of CBX8 in LC progression are not fully understood. This study aimed to investigate the regulatory role of CBX8 in LC progression. METHOD: Bioinformatics was used to analyze the relationship between CBX8 level and tumor and the enrichment pathway of CBX8 enrichment. qRT-PCR was used to detect the differential expression of CBX8 in LC cells and normal lung epithelial cells. The effects of knockdown or overexpression of CBX8 on the proliferation, migration and invasion of LC cells were evaluated by CCK- -8 assay and Transwell assay, and the levels of proteins associated with the EMT pathway and Wnt/ß-catenin signaling pathway were detected by western blot. RESULT: Bioinformatics analysis revealed that CBX8 was highly expressed in LC and enriched on the Wnt/ß-catenin signaling pathway. The expression level of CBX8 was significantly elevated in LC cells. Knockdown of CBX8 significantly inhibited cell proliferation, migration and invasion, and decreased the expression levels of EMT-related proteins and Wnt/ß-catenin pathway-related proteins. Conversely, overexpression of CBX8 promoted cell proliferation, migration and invasion, and increased the expression levels of EMT-related proteins and Wnt/ß-catenin pathway-related proteins. The Wnt inhibitor IWP-4 alleviated the effects produced by overexpression of CBX8. CONCLUSION: Collectively, these data demonstrated that CBX8 induced EMT through Wnt/ß-- catenin signaling, driving migration and invasion of LC cells.

6.
Discov Med ; 36(180): 82-90, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38273748

RESUMO

BACKGROUND: X-ray repair cross complementing 1 (XRCC1) rs1799782 polymorphism is associated with an increased risk of lung cancer (LC). The aim of this study is to analyze the underlying biological mechanisms. METHODS: Dual luciferase reporter assay was utilized to verify the impact of XRCC1 polymorphism upon promoter activity of XRCC1. Cell counting kit-8 (CCK-8) assay, colony formation assay, senescence-associated beta-galactosidase (SA-ß-gal) staining, and immunofluorescent staining were used to assess the viability, proliferation, senescence, and DNA damage of LC cells. Senescence-related proteins (cyclin dependent kinase inhibitor 1A (P21) and eukaryotic translation elongation factor 1-alpha (EF1A)) were quantified by Western blot. Chromatin immunoprecipitation was applied to validate the binding affinity of forkhead box A1 (FOXA1) and XRCC1. FOXA1-specific short hairpin RNA (shFOXA1) was used to perform the rescue assay. RESULTS: In LC cells, XRCC1 rs1799782 promoted viability and proliferation, inhibited senescence, and resulted in upregulation of EF1A as well as downregulation of P21 and phosphorylated H2A.X variant histone (γH2AX). XRCC1 rs1799782 promoted FOXA1-mediated transcription of XRCC1 through enhancing its binding to FOXA1. shFOXA1 counteracted the effects of XRCC1 rs1799782 upon the viability, proliferation, and senescence of LC cells. CONCLUSIONS: XRCC1 rs1799782 promotes DNA damage repair in LC cells through enhancing its binding to FOXA1, which facilitates FOXA1-mediated transcription of XRCC1.


Assuntos
Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/genética , Proteínas de Ligação a DNA/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/genética , Polimorfismo Genético , Dano ao DNA , Reparo do DNA/genética , Fator 3-alfa Nuclear de Hepatócito/genética
7.
Insights Imaging ; 14(1): 174, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37840068

RESUMO

BACKGROUND: Preoperative stratification is critical for the management of patients with esophageal cancer (EC). To investigate the feasibility and accuracy of PET-CT-based radiomics in preoperative prediction of clinical and pathological stages for patients with EC. METHODS: Histologically confirmed 100 EC patients with preoperative PET-CT images were enrolled retrospectively and randomly divided into training and validation cohorts at a ratio of 7:3. The maximum relevance minimum redundancy (mRMR) was applied to select optimal radiomics features from PET, CT, and fused PET-CT images, respectively. Logistic regression (LR) was applied to classify the T stage (T1,2 vs. T3,4), lymph node metastasis (LNM) (LNM(-) vs. LNM(+)), and pathological state (pstage) (I-II vs. III-IV) with features from CT (CT_LR_Score), PET (PET_LR_Score), fused PET/CT (Fused_LR_Score), and combined CT and PET features (CT + PET_LR_Score), respectively. RESULTS: Seven, 10, and 7 CT features; 7, 8, and 7 PET features; and 3, 6, and 3 fused PET/CT features were selected using mRMR for the prediction of T stage, LNM, and pstage, respectively. The area under curves (AUCs) for T stage, LNM, and pstage prediction in the validation cohorts were 0.846, 0.756, 0.665, and 0.815; 0.769, 0.760, 0.665, and 0.824; and 0.727, 0.785, 0.689, and 0.837 for models of CT_LR_Score, PET_ LR_Score, Fused_ LR_Score, and CT + PET_ LR_Score, respectively. CONCLUSIONS: Accurate prediction ability was observed with combined PET and CT radiomics in the prediction of T stage, LNM, and pstage for EC patients. CRITICAL RELEVANCE STATEMENT: PET/CT radiomics is feasible and promising to stratify stages for esophageal cancer preoperatively. KEY POINTS: • PET-CT radiomics achieved the best performance for Node and pathological stage prediction. • CT radiomics achieved the best AUC for T stage prediction. • PET-CT radiomics is feasible and promising to stratify stages for EC preoperatively.

8.
Immun Inflamm Dis ; 11(9): e1011, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37773704

RESUMO

BACKGROUND: Fibroblast activation protein-α (FAP) and livin α are considered as cancer-associated fibroblasts (CAFs) and tumor-specific targets, respectively, for immunogenic tumor vaccines. This study is designed to decipher the antitumor effect of double-gene modified dendritic cells (DCs) on Lewis lung carcinoma (LLC). METHODS: By encoding mouse FAP cDNA and human livin α (i.e., hlivin α) cDNA into recombinant adenoviral vector (rAd), rAd-FAP, rAd-hlivin α, and rAd-FAP/hlivin α were constructed, which were then transduced into mouse DCs. LLC-bearinig mice were immunized with the infected DCs (5 × 105 cells/mouse), followed by calculation of tumor volume and survival rate. The identification of CAFs from mouse LLC as well as the determination on expressions of FAP and livin α, was accomplished by western blot. Cytotoxic T lymphocyte assay was harnessed to assess the effect of the infected DCs on inducing splenic lymphocytes to lyse CAFs. RESULTS: DCs were successfully transduced with rAd-FAP/hlivin α in vitro. FAP was highly expressed in CAFs. CAFs were positive for α-SMA and negative for CD45 and CD31. Livin α level was upregulated in mouse LLC. Immunization with rAd-FAP/hlivin α-transduced DCs suppressed LLC volume and improved the survival of tumor-bearing mice. Immunization with rAd-FAP/hlivin α-transduced DCs enhanced the cytotoxic effect of splenic lymphocytes on LLC tumor-derived CAFs. CONCLUSION: Injection with rAd-FAP/hlivin α-transduced DCs promotes immune-enhanced tumor microenvironment by decreasing CAFs and suppresses tumor growth in LLC mouse models.


Assuntos
Carcinoma Pulmonar de Lewis , Animais , Humanos , Camundongos , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/terapia , Células Dendríticas , DNA Complementar/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo
9.
Food Sci Nutr ; 11(9): 5091-5101, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37701206

RESUMO

Curcumin is a polyphenol extracted from the rhizome of turmeric, and our previous research showed that curcumin inhibited cholesterol absorption and had cholesterol-lowering effect. Bisphenol A (BPA), a common plasticizer, is widely used in the manufacture of food packaging and is associated with non-alcoholic fatty liver disease (NAFLD). We hypothesized that curcumin could protect against BPA-induced hepatic steatosis by inhibiting cholesterol absorption and synthesis. Male CD-1 mice fed BPA-contaminated diet with or without curcumin for 24 weeks were used to test our hypothesis. We found that chronic low-dose BPA exposure significantly increased the levels of serum triglyceride (TG), total cholesterol (TC), and low-density lipoprotein cholesterol and the contents of liver TG and TC, resulting in liver fat accumulation and hepatic steatosis while curcumin supplementation could alleviate BPA-induced dyslipidemia and hepatic steatosis. Moreover, the anti-steatosis and cholesterol-lowering effects of curcumin against BPA coincided with a significant reduction in intestinal cholesterol absorption and liver cholesterol synthesis, which was modulated by suppressing the expression of sterol regulatory element-binding protein-2 (SREBP-2), Niemann-Pick C1-like 1 (NPC1L1), and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) in the small intestine and liver. In addition, the expression levels of liver lipogenic genes such as liver X receptor alpha (LXRα), SREBP-1c, acetyl-CoA carboxylase 1 (ACC1), and ACC2 were also markedly down-regulated by curcumin. Overall, our findings indicated that curcumin inhibited BPA-induced intestinal cholesterol absorption and liver cholesterol synthesis by suppressing SREBP-2, NPC1L1, and HMGCR expression, subsequently reducing liver cholesterol accumulation and fat synthesis, thereby preventing hepatic steatosis and NAFLD.

10.
Diagn Cytopathol ; 51(3): 182-190, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36422056

RESUMO

INTRODUCTION: Cytopathology is an important part of pathology that is used to diagnose disease on the cellular level. The application of the cell block (CB) technique plays a vital role in cytological diagnosis, as blocks and slides can be further used for special stains, immunohistochemistry (IHC), and molecular pathological analysis. Several methods for making CBs have been reported, but their procedures and cellular yield are still deemed unsatisfactory. In this article, we used gellan gum (GG) as an adjuvant for CBs, which resulted in higher cellular yield with simpler procedures. METHODS: CBs were prepared by using GG, copper sulfate, plasma/thrombin, or pregelatinized starch methods. The procedures of each of these four methods were then compared. CB sections were stained with hematoxylin and eosin (H&E), and the background and morphological features seen by H&E staining were compared. A preliminary IHC and fluorescence in situ hybridization (FISH) study was performed using cytology specimens from eleven and five cases, respectively. The expression of immunocomplex by IHC and the molecular signals detected by FISH were compared in CB sections made by the four methods and a section derived from the biopsy specimen block from the same patient. Feulgen staining, Alcian blue staining, and Masson trichrome staining were performed on the CB sections from 3 cases of pleural fluid. The cellular yield of CB sections from 83 cases according to the four methods was compared using NDP analysis software. RESULTS: The results demonstrated that sections derived from CBs made with GG had a clear background and good morphological features by H&E staining. The expression of immunocomplex by IHC and the molecular signals of FISH detection in the sections from CBs made by GG were accurately located just as those in biopsy sections from the same patient. The DNA, acidic mucus, and fibrin could be clearly identified through special stains in the CB sections. The procedures involved in the GG method were easily controllable and the coagulated gel increased the ease by which the CB was embedded and sectioned. Specifically, sections from CBs made by the GG method contained higher cellular yield because cells could be concentrated on the bottom of the gel after centrifugation. CONCLUSION: This novel method for making CBs is a practical, simple method that can result in higher cellular yield. This method is therefore worth promoting in clinical applications.


Assuntos
Citodiagnóstico , Humanos , Citodiagnóstico/métodos , Hibridização in Situ Fluorescente , Imuno-Histoquímica , Biópsia
11.
Pharmacology ; 108(1): 8-16, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36470213

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) accounts for about 85% of generally reported lung cancer patients. OBJECTIVES: This is a systematic review of the clinical efficacy and safety of osimertinib in treating epidermal growth factor receptor (EGFR) mutation-positive advanced NSCLC. METHODS: A network search was completed for clinical research literature (from inception of each database to May 30, 2020) on osimertinib for EGFR mutation-positive advanced NSCLC. Strict inclusion and exclusion criteria were formulated to screen the literature. After data extraction, RevMan 5.3 software was utilized for quality evaluation and meta-analysis. The primary endpoints were objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and adverse events of grades 3 and 4. RESULTS: Finally, 6 eligible articles and a total of 1,848 patients containing 1,123 in experimental groups and 725 in control groups were included. Meta-analysis indicated that ORR (odds ratio [OR] = 3.40, 95% CI 1.64∼7.01, p = 0.0009), DCR (OR = 4.36, 95% CI 3.09∼6.15, p < 0.00001), PFS (HR = 0.36, 95% CI 0.27∼0.47, p < 0.00001), and OS (OR = 0.58, 95% CI 0.46∼0.72, p < 0.00001) of the experimental group were prominently better than the control group. Adverse events of grades 3 and 4 mainly incorporated decreased nausea, rash, stomatitis, and vomiting, which were dramatically relieved compared with the control group. CONCLUSION: Osimertinib is currently an appreciably effective and well-tolerated therapeutic avenue for EGFR mutation-positive advanced NSCLC.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Antineoplásicos/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Compostos de Anilina/efeitos adversos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Mutação
12.
Cancer Biother Radiopharm ; 38(5): 322-335, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33769841

RESUMO

Background: Mesenchymal stem cells (MSCs) could inhibit the proliferation of lung cancer cells. The authors' study investigated the effects of immunologically activated human umbilical cord (HUC)-MSCs on A549 lung cancer cells. Materials and Methods: HUC-MSCs were separated from the umbilical cord using the adherence method. Surface markers of HUC-MSCs were detected by flow cytometry for MSC identification. Imiquimod (TLR7 agonist) was incubated with HUC-MSCs for immune activation, and the expression of MSC-specific markers and immune inflammatory molecules was measured by quantitative real-time polymerase chain reaction. HUC A549 cells were cocultured with HUC-MSCs treated with imiquimod, siTLR7 (small interfering RNA for TLR7) or TLR7 overexpression, and then cell viability, proliferation, migration, and invasion, and the expression of phosphatidylinositol-3-kinase (PI3K)/Akt and NF-κB was investigated using MTT assay, clone formation assay, transwell assay, and Western blot, respectively. Results: HUC-MSCs were identified as positive for CD73, CD105, CD44, CD29, and CD90. Expression of MSC markers was inhibited, while those of immune inflammatory molecules expression except IL-6 (interleukin-6) was enhanced after MSCs were immunologically activated by imiquimod. After being cocultured with HUC-MSCs treated with imiquimod or overexpressed TLR7, cell viability, proliferation, and metastasis, and the phosphorylation of P65 and AKT in A549 cells were decreased, but apoptosis was increased, while siTLR7 showed the opposite effect HUC. Conclusions: Immunologically activated HUC-MSCs inhibited the growth and metastasis, yet, promoted the apoptosis of A549 lung cancer cells via regulating the PI3K/Akt and NF-κB pathways.


Assuntos
Neoplasias Pulmonares , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Diferenciação Celular , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Imiquimode/farmacologia , Imiquimode/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor 7 Toll-Like/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neoplasias Pulmonares/metabolismo
13.
Technol Cancer Res Treat ; 21: 15330338221136724, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36380607

RESUMO

Background: This study is a retrospective study. The purpose of this study is to construct and validate an early warning model of lung cancer through machine learning. Methods: The CDKN2A gene expression profile and clinical information were downloaded from The Cancer Genome Atlas (TCGA) database and divided into a tumor group and a normal group (n = 57). The top 5 somatic mutation-related genes were extracted from 567 somatic mutation data downloaded from TCGA database using random forest algorithm. Cox proportional hazard model and nomogram were constructed combining CDKN2A, 5 somatic mutation-related genes, gender, and smoking index. Patients were divided into high-risk and low-risk groups according to risk score. The predictability of the model in the prognosis of lung cancer was estimated by Kaplan-Meier survival analysis and receiver operating characteristics curve. Results: We constructed a prognostic model consisting of 5 somatic mutation-related genes (sphingosine 1-phosphate receptor 1 [S1PR1], dedicator of cytokinesis 7 [DOCK7], DEAD-box helicase 4 [DDX4], laminin subunit beta 3 [LAMB3], and importin 5 [IPO5]), cyclin-dependent kinase inhibitor 2A (CDKN2A), gender, and smoking indicators. The high-risk group had a lower overall survival rate compared to the low-risk group (hazard ratio = 2.14, P = 0 .0323). The area under the curve predicted for 3-year, 5-year, and 10-year survival rates are 0.609, 0.673, and 0.698, respectively. The accuracy, sensitivity, and specificity of the model for predicting the 10-year survival rate of lung cancer are 76.19%, 56.71%, and 86.23%. Conclusion: The lung cancer early warning model and nomogram may provide an essential reference for patients with lung cancer management in the clinic.


Assuntos
Neoplasias Pulmonares , Humanos , Estudos Retrospectivos , Neoplasias Pulmonares/patologia , Prognóstico , Aprendizado de Máquina , Modelos de Riscos Proporcionais , beta Carioferinas
14.
Nutrients ; 14(9)2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35565795

RESUMO

Curcumin is a polyphenol that has been shown to have prebiotic and cholesterol-lowering properties. This study aimed to investigate the impact of curcumin on bile cholesterol supersaturation and the potential mechanistic role of intestinal microbiota and cholesterol absorption. Male hamsters (n = 8) were fed a high-fat diet (HFD) supplemented with or without curcumin for 12 weeks. Results showed that curcumin significantly decreased cholesterol levels in the serum (from 5.10 to 4.10 mmol/L) and liver (from 64.60 to 47.72 nmol/mg protein) in HFD-fed hamsters and reduced the bile cholesterol saturation index (CSI) from 1.64 to 1.08 due to the beneficial modifications in the concentration of total bile acids (BAs), phospholipids and cholesterol (p < 0.05). Gut microbiota analysis via 16S rRNA sequencing revealed that curcumin modulated gut microbiota, predominantly increasing microbiota associated with BA metabolism and short-chain fatty acid production, which subsequently up-regulated the expression of hepatic cholesterol 7-alpha hydroxylase and increased the synthesis of bile acids (p < 0.05). Furthermore, curcumin significantly down-regulated the expression of intestinal Niemann−Pick C1-like protein 1(NPC1L1) in hamsters and reduced cholesterol absorption in Caco-2 cells (p < 0.05). Our results demonstrate that dietary curcumin has the potential to prevent bile cholesterol supersaturation through modulating the gut microbiota and inhibiting intestinal cholesterol absorption.


Assuntos
Curcumina , Microbioma Gastrointestinal , Animais , Bile/metabolismo , Ácidos e Sais Biliares/metabolismo , Células CACO-2 , Colesterol , Colesterol 7-alfa-Hidroxilase/metabolismo , Cricetinae , Curcumina/metabolismo , Curcumina/farmacologia , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Humanos , Fígado/metabolismo , Masculino , RNA Ribossômico 16S/metabolismo
15.
J Recept Signal Transduct Res ; 42(3): 215-224, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33719846

RESUMO

Baicalin plays important roles in different types of cancer. A previous report showed that baicalin attenuates cisplatin resistance in lung cancer. However, its mechanism remains unclear. In this study, we investigated the effect and mechanism of baicalin on DNA repair and sensitivity of lung cancer cells to cisplatin. A549 and A549/DPP cells were treated with baicalin and cisplatin. A549/DPP cells were transfected with XRCC1 and siXRCC1. Cell viability and DNA damage were detected by MTT and comet assay. Apoptosis rate and cell cycle were detected by flow cytometry assay. The expressions of Bax, Bcl-2, and Cyclin D1 were detected by western blot. XRCC1 expression was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot. Baicalin and cisplatin decreased cell viability in A549 and A549/DPP cells in dose-dependent manner. Baicalin enhanced the effect of cisplatin on promoting apoptosis, arresting cell on S stage and triggering DNA damage accompanied with the upregulation of Bcl-2-associated X protein (Bax) and downregulation of B-cell lymphoma 2 (Bcl-2) and Cyclin D1 in A549/DPP cells. Moreover, baicalin promoted the inhibitory effect of cisplatin on XRCC1 expression in A549 and A549/DPP cells. However, the synthetic effects of baicalin and cisplatin on A549/DPP cells were partially inhibited by XRCC1 overexpression and promoted by XRCC1 knockdown. This study demonstrates that baicalin interferes with XRCC1-mediated cellar DNA repair to sensitize lung cancer cells to cisplatin.


Assuntos
Antineoplásicos , Flavonoides , Neoplasias Pulmonares , Proteína 1 Complementadora Cruzada de Reparo de Raio-X , Células A549 , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Cisplatino/farmacologia , Ciclina D1/genética , Reparo do DNA , Resistencia a Medicamentos Antineoplásicos/genética , Flavonoides/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
16.
Cancer Gene Ther ; 29(1): 101-121, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33500536

RESUMO

Significantly high-expressed circFLNA has been found in various cancer cell lines, but not in lung cancer. Therefore, this study aimed to explore the role of circFLNA in the progression of lung cancer. The target gene of circFLNA was determined by bioinformatics and luciferase reporter assay. Viability, proliferation, migration, and invasion of the transfected cells were detected by CCK-8, colony formation, wound-healing, and transwell assays, respectively. A mouse subcutaneous xenotransplanted tumor model was established, and the expressions of circFLNA, miR-486-3p, XRCC1, CYP1A1, and related genes in the cancer cells and tissues were detected by RT-qPCR, Western blot, or immunohistochemistry. The current study found that miR-486-3p was low-expressed in lung cancer. MiR-486-3p, which has been found to target XRCC1 and CYP1A1, was regulated by circFLNA. CircFLNA was located in the cytoplasm and had a high expression in lung cancer cells. Cancer cell viability, proliferation, migration, and invasion were promoted by overexpressed circFLNA, XRCC1, and CYP1A1 but inhibited by miR-486-3p mimic and circFLNA knockdown. The weight of the xenotransplanted tumor was increased by circFLNA overexpression yet reduced by miR-486-3p mimic. Furthermore, miR-486-3p mimic reversed the effect of circFLNA overexpression on promoting lung cancer cells and tumors and regulating the expressions of miR-486-3p, XRCC1, CYP1A1, and metastasis/apoptosis/proliferation-related factors. However, overexpressed XRCC1 and CYP1A1 reversed the inhibitory effect of miR-486-3p mimic on cancer cells and tumors. In conclusion, circFLNA acted as a sponge of miR-486-3p to promote the proliferation, migration, and invasion of lung cancer cells in vitro and in vivo by regulating XRCC1 and CYP1A1.


Assuntos
Neoplasias Pulmonares , MicroRNAs , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Filaminas/genética , Filaminas/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Camundongos , MicroRNAs/metabolismo , RNA Circular/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/metabolismo
17.
Lancet Gastroenterol Hepatol ; 6(9): 700-708, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34297944

RESUMO

BACKGROUND: White light endoscopy is a pivotal first-line tool for the detection of gastric neoplasms. However, gastric neoplasms can be missed during upper gastrointestinal endoscopy due to the subtle nature of these lesions and varying skill among endoscopists. Here, we aimed to evaluate the effect of an artificial intelligence (AI) system designed to detect focal lesions and diagnose gastric neoplasms on reducing the miss rate of gastric neoplasms in clinical practice. METHODS: This single-centre, randomised controlled, tandem trial was done at Renmin Hospital of Wuhan University, China. We recruited consecutive patients (≥18 years old) undergoing routine upper gastrointestinal endoscopy for screening, surveillance, or investigation of symptoms. Same-day tandem upper gastrointestinal endoscopy was done where patients first underwent either AI-assisted (AI-first) or routine (routine-first) white light endoscopy, followed immediately by the other procedure, with targeted biopsies for all detected lesions taken at the end of the second examination. Patients were randomly assigned (1:1) to the AI-first or routine-first group using a computer-generated random numerical series and block randomisation (block size of four). Endoscopists were not blinded to randomisation status, whereas patients and pathologists were. The primary endpoint was the miss rate of gastric neoplasms and the analysis was done per protocol. This trial is registered with the Chinese Clinical Trial Registry, ChiCTR2000034453, and has been completed. FINDINGS: Between July 6, 2020, and Dec 11, 2020, 907 patients were randomly assigned to the AI-first group and 905 to the routine-first group. The gastric neoplasm miss rate was significantly lower in the AI-first group than in the routine-first group (6·1%, 95% CI 1·6-17·9 [3/49] vs 27·3%, 15·5-43·0 [12/44]; relative risk 0·224, 95% CI 0·068-0·744; p=0·015). The only reported adverse event was bleeding from a target lesion after biopsy. INTERPRETATION: The use of an AI system during upper gastrointestinal endoscopy significantly reduced the gastric neoplasm miss rate. AI-assisted endoscopy has the potential to improve the yield of gastric neoplasms by endoscopists. FUNDING: The Project of Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision and the Hubei Province Major Science and Technology Innovation Project.


Assuntos
Inteligência Artificial , Aprendizado Profundo , Endoscopia do Sistema Digestório/métodos , Programas de Rastreamento/métodos , Neoplasias Gástricas/diagnóstico , Adulto , China/epidemiologia , Feminino , Seguimentos , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Neoplasias Gástricas/epidemiologia
18.
Open Med (Wars) ; 16(1): 183-191, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33585694

RESUMO

OBJECTIVE: The aim of the present work was to investigate the clinical efficacy of first-line chemotherapy regimens in the treatment of advanced non-small cell lung cancer (NSCLC) through a comprehensive network meta-analysis (NMA). METHODS: The prospective randomized controlled clinical trials relevant to 10 first-line chemotherapy regimens in the treatment of advanced NSCLC were systematic electronic search in the databases of Pubmed, Embase, Cochrane Library and CNKI. The combined direct or indirect objective response rate (ORR) between each of the 10 first-line chemotherapy regimens was calculated. RESULTS: Seventeen prospective clinical trials of first-line chemotherapy regimens in treatment of advanced NSCLC were included in the NMA. The 10 treatment regimens including A = cisplatin + gemcitabine, B = carboplatin + gemcitabine, C = gemcitabine, D = carboplatin + paclitaxel, E = paclitaxel + gemcitabine, F = docetaxel + carboplatin, G = gemcitabine + vinorelbine, H = pemetrexed + carboplatin, I = cisplatin + pemetrexed and J = cisplatin + docetaxel were compared in the present NMA. Direct pooled results indicated that the ORR was not statistically different (P all > 0.05). However, NMA showed that the combined ORR for regimens A (OR = 1.47, 95% CI: 0.80-2.81), B (OR = 3.22, 95% CI: 1.45-6.923), D (OR = 3.30, 95% CI: 1.22-9.33), E (OR = 4.36, 95% CI: 1.64-12.82), G (OR = 3.72, 95% CI: 1.12-12.83) and I (OR = 5.80, 95% CI: 2.04-17.86) was superior to regimen C. Rank probability analysis indicated that regimen C = gemcitabine and regimen I = cisplatin + pemetrexed had the highest probability of inferior and superior treatment ORR among the 10 first-line chemotherapy regimens. CONCLUSION: Cisplatin + pemetrexed may have particularly prominent ORR for advanced NSCLC as the first-line chemotherapy regimen.

19.
Thorac Cancer ; 11(2): 329-335, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31846184

RESUMO

BACKGROUND: To investigate the diagnostic efficacy of long noncoding RNA metastasis-associated in lung adenocarcinoma transcript l (MALAT1) as a candidate serological biomarker for non-small cell lung cancer (NSCLC). METHODS: Diagnostic studies relevant to circulation long noncoding RNA MALAT1 as a candidate serological biomarker for NSCLC were electronically systematically searched in PubMed, EMBASE, EBSCO and CNKI databases. Suitable studies were included in the meta-analysis by pooling the diagnostic sensitivity, specificity, positive likelihood ratio (+LR), negative likelihood ratio (-LR), diagnostic odds ratio (DOR) and area under the symmetric ROC curve (AUC) through a random or fixed effects model. Deeks' funnel plot was applied for publication bias evaluation. RESULTS: Six studies with eight datasets were finally included in the meta-analysis after a systematic search of the databases was performed. The pooled diagnostic sensitivity, specificity, +LR, -LR and DOR were 0.81 (95% CI:0.78-0.84), 0.67 (95% CI:0.63-0.71), 2.61 (95% CI:1.81-3.71), 0.28 (95% CI:0.19-0.43) and 13.73 (95% CI:6.19-30.44), respectively. The pooled area under the ROC curve (AUC) were 0.8663 and 0.8658, respectively by symmetric and asymmetric methods. CONCLUSION: Based on the results of our study, serum long noncoding RNA MALAT1 is a promising biomarker for NSCLC screening. However, due to its low specificity, MALAT1 positive cases need further validation for NSCLC by other diagnostic methods such as radiology, cytology, etc.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Neoplasias Pulmonares/diagnóstico , RNA Longo não Codificante/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/genética , Prognóstico
20.
Aging (Albany NY) ; 11(7): 2138-2150, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30981205

RESUMO

Non-small cell lung cancer (NSCLC) is the most common and lethal human malignant tumor worldwide. Platinum-based chemotherapy is still the mainstay of treatment for NSCLC. However, long-term chemotherapy usually induces serious drug resistance in NSCLC cells. Accordingly, treatment strategies that reverse the resistance of NSCLC cells against platinum-based drugs may have considerable clinical value. In the present study, we observed significant upregulation of CAV-1 expression and a significant decrease of miR-204 expression in cisplatin-resistant A549 (CR-A549) and cisplatin-resistant PC9 (CR-PC9) cells compared to their parental A549 and PC9 cells. Furthermore, we demonstrated that the downregulation of miR-204 expression was responsible for CAV-1 overexpression in these cisplatin-resistant NSCLC cells. We then found that enforced expression of miR-204 can resensitize CR-A549 and CR-PC9 cells to cisplatin-induced mitochondrial apoptosis through suppression of the caveolin-1/AKT/Bad pathway. We demonstrated that dysregulation of miR-204/caveolin-1 axis is an important mechanism for NSCLC cells to develop the chemoresistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Caveolina 1/genética , Cisplatino/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , MicroRNAs/genética , Células A549 , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Nus , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína de Morte Celular Associada a bcl/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA