Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Int J Mol Med ; 53(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38186308

ABSTRACT

Disulfiram (DSF) is used to treat non­small cell lung cancer (NSCLC). DSF significantly increases expression of programmed death­ligand 1 (PD­L1), which may enhance immunosuppression and immune escape of tumors. Therefore, the present study aimed to investigate the role of combined treatment of DSF and anti­PD­L1 in NSCLC resistance. The viability and apoptosis of A549 cells were detected by the Cell Counting Kit­8 assay and flow cytometry, respectively. The expression levels of ATPase copper­transporting ß (ATP7B) and PD­L1 in A549 cells were detected by reverse transcription­quantitative PCR and western blot analysis. The levels of reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) in A549 cells were detected by respective assay kits. The expression levels of cuproptosis­associated proteins ferredoxin­1 (FDX1), ATP7B, solute carrier family 31 member 1 (SLC31A1), succinate dehydrogenase B (SDHB), PD­L1 and hypoxia inducible factor (HIF)­1A were analyzed by western blotting in A549 cells. DSF inhibited the viability of A549 cells and promoted expression levels of ATP7B and PD­L1 at both mRNA and protein levels in A549 cells. The viability of cisplatin (DPP)­treated A549 cells was increased following DSF treatment. JQ­1 (a PD­L1 inhibitor) suppressed the viability of DPP­treated A549 cells pretreated with DSF. DSF increased expression levels of ATP7B and PD­L1. The combination treatment of DSF and JQ­1 in A549 cells increased levels of ROS and MDA, as well as expression levels of FDX1 and SLC31A1; however, combination treatment decreased levels of SOD, as well as expression levels of ATP7B, SDHB, PD­L1, and HIF­1A. PX478 (an HIF­1 inhibitor) acted with DSF to enhance the inhibitory effects on the viability and on the induction of apoptosis of A549 cells. PX478 upregulated the levels of ROS and MDA, while it downregulated levels of SOD in DSF­treated A549 cells. PX478 promoted expression levels of FDX1 and SLC31A1, while it suppressed expression levels of ATP7B, PD­L1, and HIF­1A in DSF­treated A549 cells. In conclusion, the combined treatment of A549 cells with anti­PD­L1 and DSF enhanced the effect of cuproptosis on the inhibition of NSCLC cell viability.


Subject(s)
3,4-Methylenedioxyamphetamine , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , B7-H1 Antigen/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Disulfiram/pharmacology , Reactive Oxygen Species , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Signal Transduction , Superoxide Dismutase
2.
Epigenomics ; 15(4): 195-207, 2023 02.
Article in English | MEDLINE | ID: mdl-37125501

ABSTRACT

Aim: Possible roles of miRNAs in cancer treatment have been highly studied. This study aimed to elucidate the role of miR-4732-3p in lung cancer. Methods: Bioinformatics analysis was conducted to predict miR-4732-3p-related mRNA targets in lung cancer. Following interaction determination between miR-4732-3p and TBX15 as well as between TBX15 and TNFSF11, their in vitro and in vivo roles were assayed. Results: miR-4732-3p negatively targeted TBX15, which upregulated TNFSF11 by enhancing the activity of the TNFSF11 promoter. Overexpression of miR-4732-3p or silencing of TBX15 or TNFSF11 inhibited the malignant phenotype of lung cancer cells and reduced tumorigenicity in vivo. Conclusion: Overall, this study highlighted the inhibitory role of miR-4732-3p in lung cancer progression through the TBX15/TNFSF11 axis.


This study describes the role of miR-4732-3p in lung cancer. The authors conducted bioinformatics analysis to predict miR-4732-3p-related mRNA targets in lung cancer. Then they analyzed the potential interaction between miR-4732-3p and TBX15 and between TBX15 and TNFSF11. To evaluate their effects on the progression of lung cancer, the authors performed in vitro and in vivo assays. They discovered that miR-4732-3p negatively targeted TBX15, which upregulated TNFSF11 by enhancing the activity of TNFSF11 promoter. Overexpression of miR-4732-3p or silencing of TBX15 or TNFSF11 inhibited the malignant phenotype of lung cancer cells and reduced tumorigenicity in vivo. This study, which has identified potential for the miR-4732-3p/TBX15/TNFSF11 axis as an antioncogenic tool, opens the possibility for better monitoring of lung cancer.


Subject(s)
Lung Neoplasms , MicroRNAs , Humans , Cell Proliferation , Computational Biology , Lung Neoplasms/genetics , MicroRNAs/genetics , Phenotype , Promoter Regions, Genetic , RANK Ligand , T-Box Domain Proteins/genetics
3.
Dis Markers ; 2022: 7098463, 2022.
Article in English | MEDLINE | ID: mdl-36204510

ABSTRACT

Background: Lung fibrosis is a severe lung disorder featured by chronic nonspecific inflammation of the interstitial lung and deposition of collagen, leading to lung dysfunction. It has been identified that ferroptosis is involved in the progression of lung injury. Particulate matter (PM2.5) is reported to be correlated with the incidence of pulmonary fibrosis. However, mechanisms underlying ferroptosis in PM2.5-related lung fibrosis is unclear. In this study, we aimed to explore the effect of PM2.5 on ferroptosis in lung fibrosis and the related molecular mechanisms. Methods: PM2.5-treated mouse model and cell model were established. Fibrosis and tissue damage were measured by Masson's trichrome staining and HE staining. Fibrosis biomarkers, such as α-SMA, collagen I, and collagen III, were examined by histological analysis. The ferroptosis phenotypes, including the levels of iron, Fe2+, MDA, and GSH, were measured by commercial kits. ROS generation was checked by DCFH-DA. The oxidative stress indicators, 3-nitro-L-tyrosine (3'-NT), 4-HNE, and protein carbonyl, were checked by enzyme linked immunosorbent assay (ELISA). The thiobarbituric acid reactive substances (TBARS) and GSH/GSSG ratio were assessed by TBARS assay kit and GSH/GSSG assay kit, respectively. TGF-ß signaling was detected by Western blotting. Results: PM2.5 induced the lung injury and fibrosis in the mice model, along with elevated expression of fibrosis markers. PM2.5 enhanced oxidative stress in the lung of the mice. The SOD2 expression was reduced, and NRF2 expression was enhanced in the mice by the treatment with PM2.5. PM2.5 triggered ferroptosis, manifested as suppressed expression of GPX4 and SLC7A11, decreased levels of iron, Fe2+, and MDA, and increased GSH level in mouse model and cell model. The TGF-ß and Smad3 signaling was inhibited by PM2.5. ROS inhibitor NAC reversed PM2.5-regulated ROS and ferroptosis in primary mouse lung epithelial cells. Conclusions: Therefore, we concluded that PM2.5 exposure induced lung injury and fibrosis by inducing ferroptosis via TGF-ß signaling.


Subject(s)
Ferroptosis , Lung Injury , Pulmonary Fibrosis , Animals , Collagen Type I , Fibrosis , Glutathione Disulfide , Iron , Lung Injury/chemically induced , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Particulate Matter/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Reactive Oxygen Species , Thiobarbituric Acid Reactive Substances , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
Environ Pollut ; 272: 116024, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33187840

ABSTRACT

We collected 170 samples of airborne fine particulate matter from five coal-producing cities and one oil-producing city in northern China during both heating and non-heating periods to quantify the concentrations of 12 polycyclic aromatic hydrocarbons, estimate their bioaccessible fraction, and calculate the incremental lifetime cancer risk (ILCR) of this fraction. The major sources of the particulate matter were analyzed using the chemical mass balance model. We found that the main emission sources were coal combustion during the heating period and open sources during the non-heating period. The ILCR was initially calculated as 2.65 × 10-9 for coal-producing cities and 4.60 × 10-9 for the oil-producing city during the heating period and 1.17 × 10-8 and 3.34 × 10-8, respectively, during the non-heating period. When only the bioaccessible fraction was used, the ILCR in coal-producing cities and the oil-producing city decreased by 87.2% and 82.1%, respectively, for the heating period and by 89.0% and 80.1%, respectively, for the non-heating period. The findings suggest that bioaccessibility should be considered when assessing the carcinogenic risk of polycyclic aromatic hydrocarbons. This study provides insights into the contribution of major emission sources to air pollution related to the long-term exploitation, transportation, and use of coal and oil.


Subject(s)
Air Pollutants , Polycyclic Aromatic Hydrocarbons , Air Pollutants/analysis , China , Cities , Coal/analysis , Environmental Monitoring , Particulate Matter/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Risk Assessment
5.
Front Immunol ; 11: 366, 2020.
Article in English | MEDLINE | ID: mdl-32194569

ABSTRACT

Patients with triple-negative breast cancer (TNBC) suffer an unfavorable prognosis. Carboplatin (CBDCA) as a cytotoxic reagent has been widely administered to patients with cancer including TNBC. Programmed cell death protein 1 (PD-1) is an immune checkpoint, blockade of which unleashes T cell functions that kill cancer cells. However, the efficacy of CBDCA combined with anti-PD-1 antibodies in TNBC has not been determined. Patient-derived xenografts (PDX) were implanted to immune-deficient mice. Three mouse TNBC cell lines (4T1, EMT6, and E0771) were seeded to immune-competent mice. Tumor volumes and survival rates were monitored. CBDCA and anti-PD-1 antibodies were administered by intra-peritoneal injection at designated time points. Total CD8+ T cells, memory CD8+ T cells, and CD103+ dendritic cells (DC) in the tumor were measured by flow cytometry. Tumor-specific CD8+ T cells were quantified by the ELISpot assay. Administration of CBDCA to PDX-bearing mice induced increased levels of tumor cell necrosis and reduced tumor size. Treatment with CBDCA and anti-PD-1 antibodies reduced TNBC tumor volumes and slightly improved survival rates. More importantly, therapy with CBDCA and anti-PD-1 antibodies before surgery showed a remarkably improved, sustainable protection against a secondary tumor after surgery by a CD8+- T-cell-dependent manner, which required CCL4 expressed in the tumor and subsequently CD103+ DC recruited to the tumor microenvironment. Immunochemotherapy with CBDCA and anti-PD-1 antibodies before surgery improves the outcome of a secondary tumor after surgery via increasing the number of tumor-specific CD8+ T cells in the tumor microenvironment of murine TNBC. These results highlight the possibility to utilize this regimen in clinical practice.


Subject(s)
Carboplatin/administration & dosage , Immune Checkpoint Inhibitors/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Animals , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL4/physiology , Dendritic Cells/immunology , Female , Humans , Mice , Middle Aged , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
J Cancer ; 10(22): 5377-5387, 2019.
Article in English | MEDLINE | ID: mdl-31632482

ABSTRACT

Tumor-associated macrophages (TAM) have been shown to support tumor growth and progression by various mechanisms. However, the roles of TAM in gastric cancer (GC) peritoneal metastasis remain elusive. To explore the roles of macrophages in the process of GC peritoneal metastasis, we performed the present study. Samples from the primary GC tumor beds, surgical margins, peritoneal metastatic lesions and surrounding tissue, and the Pouch of Douglas, were collected, fixed by formalin, and embedded with paraffin. Immunohistochemistry staining for macrophages markers was performed. The peritoneal lavage was obtained from a fraction of patients to analyze the ratios of epidermal growth factor (EGF)- and vascular endothelial growth factor (VEGF)-secreting macrophages in the peritoneal cavity. GC patients with peritoneal metastasis had increased levels of macrophages and alternatively activated macrophages in the peritoneum compared to those without dissemination. Patients bearing more macrophages in the peritoneum had a poorer prognosis. GC patients bearing peritoneal metastasis harbored an increased level of angiogenesis. Macrophages in the peritoneal cavity were a source of EGF and VEGF. Macrophages in the peritoneum of GC patients play a supportive role for peritoneal metastasis by producing EGF and VEGF. Macrophages in the peritoneum might be a therapeutic target in the future.

7.
Oncol Lett ; 15(3): 3287-3294, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435071

ABSTRACT

The activation of the Wnt/ß-catenin signaling pathway has been demonstrated to play important roles in breast carcinogenesis and to be associated with a poorer prognosis in breast cancer patients. However, genetic mutation is not the major reason for Wnt/ß-catenin activation in breast cancer. Dishevelled-associated antagonist of ß-catenin homolog 2 (DACT2) is a negative regulator of ß-catenin and acts as a tumor suppressor in numerous cancer types; however, the expression change and potential role of DACT2 in breast cancer is unknown. The present study detected the expression and function of DACT2 in breast cancer progression. It was identified that the expression of DACT2 significantly decreased in breast cancer tissues compared with paired adjacent normal breast tissues. Additional investigation demonstrated that the hypermethylation of DACT2 gene promoter contributes to the loss of the gene in breast cancer. It was also demonstrated that DACT2 is a tumor suppressor in breast cancer and inhibits the proliferation and invasion of breast cancer cells by repressing the expression of ß-catenin target genes associated with tumor growth and metastasis. The present study indicates that the loss of DACT2 may contribute to breast cancer progression and provides a promising therapeutic target for the treatment of breast cancer.

8.
Oncotarget ; 6(9): 7084-103, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25762626

ABSTRACT

The role of autophagy in cancers is controversial. Here we aim to determine the prognostic significance of autophagy in colorectal carcinoma patients, thereby allowing more rational development of therapeutic strategies. Through transmission electron microscopy, our data first demonstrated high frequency of defective mitochondria was strongly associated with poor overall survival in colorectal carcinoma. Next immunohistochemical study showed the expressions of Beclin 1, LC3B and Bcl-xL in both the center of tumor and adjacent noncancerous mucosal region were also correlated with overall survivals. We developed an autophagy signature for prognosis based on these three major autophagic proteins, further analysis suggested it was an independent prognostic biomarker and had its value even within single clinical stage. Combined TNM stage and this signature could significantly improve the accuracy of survival prognosis. To validate these immunohistochemical results, an internal testing cohort and an independent population were also included. Our findings suggest that autophagy plays an important role in the clinical cancer progression. Therefore autophagic proteins may be valuable prognostic biomarkers in the therapy of colorectal carcinoma and possibly other types of cancers.


Subject(s)
Autophagy , Carcinoma/drug therapy , Colorectal Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins/metabolism , Area Under Curve , Beclin-1 , Biomarkers, Tumor/metabolism , Carcinoma/diagnosis , Carcinoma/metabolism , Cell Survival , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Disease Progression , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Middle Aged , Mitochondria/pathology , Prognosis , Proportional Hazards Models , Treatment Outcome , bcl-X Protein/metabolism
9.
Med Oncol ; 29(3): 1656-62, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21928106

ABSTRACT

A platinum-based two-drug regimen is currently the standard of care for patients with advanced non-small-cell lung cancer (NSCLC). However, chemotherapy-induced side effects still remain a significant clinical problem. Astragalus polysaccharide (APS) is a polysaccharide isolated from the radix of astragalus membranaceus, a commonly used herbal compound in traditional Chinese medicine. APS was reported to increase tumor response, stabilize and improve performance status, and reduce chemotherapy toxicity. We designed this trial to determine whether APS injection integrated with vinorelbine and cisplatin (VC) offered an improved QOL over VC for patients with advanced NSCLC. Secondary objectives were tumor response, toxicity, and survival results. One hundred thirty-six patients with histologically or cytologically confirmed NSCLC were enrolled in this study from May 2008 to March 2010. Patients were randomized to receive either VC (VC arm) or VC combined with APS (VC-APS arm). The objective response rate of was 42.64% in the VC-APS arm and 36.76% in the VC arm. The difference was not statistically significant (P = 0.483). Median survival time was 10.7 and 10.2 months (P = 0.76) in VC-APS arm and VC arm, with 1-year survival rates of 35.3 and 32.4% (P = 0.717), respectively. After 3 cycles of treatment, there were significant differences in the overall patient QOL (P = 0.003), physical function (P = 0.01), fatigue (P < 0.001), nausea and vomiting (P < 0.001), pain (P = 0.007), and loss of appetite (P = 0.023) between the two study groups. In summary, we have proved that the treatment of APS integrated with VC had significantly improved QOL in patients with advanced NSCLC compared with VC alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Astragalus Plant , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Phytotherapy/methods , Polysaccharides/therapeutic use , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Astragalus Plant/chemistry , Carcinoma, Non-Small-Cell Lung/mortality , Cisplatin/administration & dosage , Cisplatin/adverse effects , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Quality of Life , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinorelbine
SELECTION OF CITATIONS
SEARCH DETAIL
...