Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cancer Med ; 13(13): e7424, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38988047

ABSTRACT

BACKGROUND: Gastric cancer (GC) is the fourth leading cause of cancer-related death worldwide. Minichromsome maintenance proteins family member 8 (MCM8) assists DNA repair and DNA replication. MCM8 exerts tumor promotor function in multiple digestive system tumors. MCM8 is also considered as a potential cancer therapeutic target. METHODS: Bioinformatics methods were used to analyze MCM8 expression and clinicopathological significance. MCM8 expression was detected by immunohistochemistry (IHC) staining and qRT-PCR. MCM8 functions in GC cell were explored by Celigo cell counting, colony formation, wound-healing, transwell, and annexin V-APC staining assays. The target of MCM8 was determined by human gene expression profile microarray. Human phospho-kinase array kit evaluated changes in key proteins after ribosomal protein S15A (RPS15A) knockdown. MCM8 functions were reassessed in xenograft mouse model. IHC detected related proteins expression in mouse tumor sections. RESULTS: MCM8 was significantly upregulated and predicted poor prognosis in GC. High expression of MCM8 was positively correlated with lymph node positive (p < 0.001), grade (p < 0.05), AJCC Stage (p < 0.001), pathologic T (p < 0.01), and pathologic N (p < 0.001). MCM8 knockdown inhibited proliferation, migration, and invasion while promoting apoptosis. RPS15A expression decreased significantly after MCM8 knockdown. It was also the only candidate target, which ranked among the top 10 downregulated differentially expressed genes (DEGs) in sh-MCM8 group. RPS15A was identified as the target of MCM8 in GC. MCM8/RPS15A promoted phosphorylation of P38α, LYN, and p70S6K. Moreover, MCM8 knockdown inhibited tumor growth, RPS15A expression, and phosphorylation of P38α, LYN, and p70S6K in vivo. CONCLUSIONS: MCM8 is an oncogene and predicts poor prognosis in GC. MCM8/RPS15A facilitates GC progression.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Ribosomal Proteins , Stomach Neoplasms , Humans , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Animals , Mice , Prognosis , Female , Male , Cell Line, Tumor , Disease Progression , Middle Aged , Minichromosome Maintenance Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , Apoptosis , Mice, Nude , Cell Movement , Xenograft Model Antitumor Assays , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
2.
J Exp Clin Cancer Res ; 43(1): 103, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570866

ABSTRACT

BACKGROUND: Brain metastasis (BM) is common among cases of advanced non-small cell lung cancer (NSCLC) and is the leading cause of death for these patients. Mesothelin (MSLN), a tumor-associated antigen expressed in many solid tumors, has been reported to be involved in the progression of multiple tumors. However, its potential involvement in BM of NSCLC and the underlying mechanism remain unknown. METHODS: The expression of MSLN was validated in clinical tissue and serum samples using immunohistochemistry and enzyme-linked immunosorbent assay. The ability of NSCLC cells to penetrate the blood-brain barrier (BBB) was examined using an in vitro Transwell model and an ex vivo multi-organ microfluidic bionic chip. Immunofluorescence staining and western blotting were used to detect the disruption of tight junctions. In vivo BBB leakiness assay was performed to assess the barrier integrity. MET expression and activation was detected by western blotting. The therapeutic efficacy of drugs targeting MSLN (anetumab) and MET (crizotinib/capmatinib) on BM was evaluated in animal studies. RESULTS: MSLN expression was significantly elevated in both serum and tumor tissue samples from NSCLC patients with BM and correlated with a poor clinical prognosis. MSLN significantly enhanced the brain metastatic abilities of NSCLC cells, especially BBB extravasation. Mechanistically, MSLN facilitated the expression and activation of MET through the c-Jun N-terminal kinase (JNK) signaling pathway, which allowed tumor cells to disrupt tight junctions and the integrity of the BBB and thereby penetrate the barrier. Drugs targeting MSLN (anetumab) and MET (crizotinib/capmatinib) effectively blocked the development of BM and prolonged the survival of mice. CONCLUSIONS: Our results demonstrate that MSLN plays a critical role in BM of NSCLC by modulating the JNK/MET signaling network and thus, provides a potential novel therapeutic target for preventing BM in NSCLC patients.


Subject(s)
Benzamides , Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Imidazoles , Lung Neoplasms , Triazines , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Mesothelin , Lung Neoplasms/pathology , GPI-Linked Proteins/metabolism , Crizotinib , Cell Line, Tumor , Brain Neoplasms/pathology
3.
Free Radic Biol Med ; 213: 150-163, 2024 03.
Article in English | MEDLINE | ID: mdl-38190923

ABSTRACT

Quercetin (Quer) is a natural flavonoid known for its inhibitory effects against various cancers. However, the mechanism by which Quer inhibits gastric cancer (GC) has not yet been fully elucidated. Ferroptosis, a mode of programmed cell death resulting from lipid peroxidation, is regulated by abnormalities in the antioxidant system and iron metabolism. Through flow cytometry and other detection methods, we found that Quer elevated lipid peroxidation levels in GC cells. Transmission electron microscopy confirmed an increase in ferroptosis in Quer-induced GC. We demonstrated that Quer inhibits SLC1A5 expression. Molecular docking revealed Quer's binding to SLC1A5 at SER-343, SER-345, ILE-423, and THR-460 residues. Using immunofluorescence and other experiments, we found that Quer altered the intracellular ROS levels, antioxidant system protein expression levels, and iron content. Mechanistically, Quer binds to SLC1A5, inhibiting the nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2), resulting in decreased xCT/GPX4 expression. Quer/SLC1A5 signaling activated p-Camk2, leading to upregulated p-DRP1 and enhanced ROS release. Additionally, Quer increased the intracellular iron content by inhibiting SLC1A5. These three changes collectively led to ferroptosis in GC cells. In conclusion, Quer targets SLC1A5 in GC cells, inhibiting the NRF2/xCT pathway, activating the p-Camk2/p-DRP1 pathway, and accelerating iron deposition. Ultimately, Quer promotes ferroptosis in GC cells, inhibiting GC progression. Overall, our study reveals that Quer can potentially impede GC progression by targeting SLC1A5, offering novel therapeutic avenues through the modulation of ferroptosis and iron homeostasis.


Subject(s)
Ferroptosis , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Quercetin/pharmacology , NF-E2-Related Factor 2/genetics , Antioxidants , Ferroptosis/genetics , Molecular Docking Simulation , Reactive Oxygen Species , Iron , Minor Histocompatibility Antigens , Amino Acid Transport System ASC
4.
Lancet Digit Health ; 5(9): e560-e570, 2023 09.
Article in English | MEDLINE | ID: mdl-37625894

ABSTRACT

BACKGROUND: Mediastinal neoplasms are typical thoracic diseases with increasing incidence in the general global population and can lead to poor prognosis. In clinical practice, the mediastinum's complex anatomic structures and intertype confusion among different mediastinal neoplasm pathologies severely hinder accurate diagnosis. To solve these difficulties, we organised a multicentre national collaboration on the basis of privacy-secured federated learning and developed CAIMEN, an efficient chest CT-based artificial intelligence (AI) mediastinal neoplasm diagnosis system. METHODS: In this multicentre cohort study, 7825 mediastinal neoplasm cases and 796 normal controls were collected from 24 centres in China to develop CAIMEN. We further enhanced CAIMEN with several novel algorithms in a multiview, knowledge-transferred, multilevel decision-making pattern. CAIMEN was tested by internal (929 cases at 15 centres), external (1216 cases at five centres and a real-world cohort of 11 162 cases), and human-AI (60 positive cases from four centres and radiologists from 15 institutions) test sets to evaluate its detection, segmentation, and classification performance. FINDINGS: In the external test experiments, the area under the receiver operating characteristic curve for detecting mediastinal neoplasms of CAIMEN was 0·973 (95% CI 0·969-0·977). In the real-world cohort, CAIMEN detected 13 false-negative cases confirmed by radiologists. The dice score for segmenting mediastinal neoplasms of CAIMEN was 0·765 (0·738-0·792). The mediastinal neoplasm classification top-1 and top-3 accuracy of CAIMEN were 0·523 (0·497-0·554) and 0·799 (0·778-0·822), respectively. In the human-AI test experiments, CAIMEN outperformed clinicians with top-1 and top-3 accuracy of 0·500 (0·383-0·633) and 0·800 (0·700-0·900), respectively. Meanwhile, with assistance from the computer aided diagnosis software based on CAIMEN, the 46 clinicians improved their average top-1 accuracy by 19·1% (0·345-0·411) and top-3 accuracy by 13·0% (0·545-0·616). INTERPRETATION: For mediastinal neoplasms, CAIMEN can produce high diagnostic accuracy and assist the diagnosis of human experts, showing its potential for clinical practice. FUNDING: National Key R&D Program of China, National Natural Science Foundation of China, and Beijing Natural Science Foundation.


Subject(s)
Mediastinal Neoplasms , Humans , Mediastinal Neoplasms/diagnosis , Mediastinum , Artificial Intelligence , Cohort Studies , Diagnosis, Computer-Assisted
5.
Lancet Digit Health ; 4(5): e309-e319, 2022 05.
Article in English | MEDLINE | ID: mdl-35341713

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) genotype is crucial for treatment decision making in lung cancer, but it can be affected by tumour heterogeneity and invasive biopsy during gene sequencing. Importantly, not all patients with an EGFR mutation have good prognosis with EGFR-tyrosine kinase inhibitors (TKIs), indicating the necessity of stratifying for EGFR-mutant genotype. In this study, we proposed a fully automated artificial intelligence system (FAIS) that mines whole-lung information from CT images to predict EGFR genotype and prognosis with EGFR-TKI treatment. METHODS: We included 18 232 patients with lung cancer with CT imaging and EGFR gene sequencing from nine cohorts in China and the USA, including a prospective cohort in an Asian population (n=891) and The Cancer Imaging Archive cohort in a White population. These cohorts were divided into thick CT group and thin CT group. The FAIS was built for predicting EGFR genotype and progression-free survival of patients receiving EGFR-TKIs, and it was evaluated by area under the curve (AUC) and Kaplan-Meier analysis. We further built two tumour-based deep learning models as comparison with the FAIS, and we explored the value of combining FAIS and clinical factors (the FAIS-C model). Additionally, we included 891 patients with 56-panel next-generation sequencing and 87 patients with RNA sequencing data to explore the biological mechanisms of FAIS. FINDINGS: FAIS achieved AUCs ranging from 0·748 to 0·813 in the six retrospective and prospective testing cohorts, outperforming the commonly used tumour-based deep learning model. Genotype predicted by the FAIS-C model was significantly associated with prognosis to EGFR-TKIs treatment (log-rank p<0·05), an important complement to gene sequencing. Moreover, we found 29 prognostic deep learning features in FAIS that were able to identify patients with an EGFR mutation at high risk of TKI resistance. These features showed strong associations with multiple genotypes (p<0·05, t test or Wilcoxon test) and gene pathways linked to drug resistance and cancer progression mechanisms. INTERPRETATION: FAIS provides a non-invasive method to detect EGFR genotype and identify patients with an EGFR mutation at high risk of TKI resistance. The superior performance of FAIS over tumour-based deep learning methods suggests that genotype and prognostic information could be obtained from the whole lung instead of only tumour tissues. FUNDING: National Natural Science Foundation of China.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Artificial Intelligence , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , ErbB Receptors/therapeutic use , Genes, erbB-1 , Genotype , Humans , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Prospective Studies , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies
6.
Br J Cancer ; 126(12): 1795-1805, 2022 06.
Article in English | MEDLINE | ID: mdl-35217799

ABSTRACT

BACKGROUND: The lack of non-invasive methods for detection of early micro-metastasis is a major cause of the poor prognosis of non-small cell lung cancer (NSCLC) brain metastasis (BM) patients. Herein, we aimed to identify circulating biomarkers based on proteomics for the early diagnosis and monitoring of patients with NSCLC BM. METHODS: Upregulated proteins were detected by secretory proteomics in the animal-derived high brain metastatic lung cancer cell line. A well-designed study composed of three independent cohorts was then performed to verify these blood-based protein biomarkers: the serum discovery and verification cohorts (n = 80; n = 459), and the tissue verification cohort (n = 76). Logistic regression was used to develop a diagnostic biomarker panel. Model validation cohort (n = 160) was used to verify the stability of the constructed predictive model. Changes in serum Cathepsin F (CTSF) levels of patients were tracked to monitor the treatment response. Progression-free survival (PFS) and overall survival (OS) were analysed to assess their prognostic relevance. RESULTS: CTSF and Fibulin-1 (FBLN1) levels were specifically upregulated in sera and tissues of patients with NSCLC BM compared with NSCLC without BM and primary brain tumour. The combined diagnostic performance of CTSF and FBLN1 was superior to their individual ones. CTSF serum changes were found to reflect the therapeutic response of patients with NSCLC BM and the trends of progression were detected earlier than the magnetic resonance imaging changes. Elevated expression of CTSF in NSCLC BM tissues was associated with poor PFS, and was found to be an independent prognostic factor. CONCLUSIONS: We report a novel blood-based biomarker panel for early diagnosis, monitoring of therapeutic response, and prognostic evaluation of patients with NSCLC BM.


Subject(s)
Brain Neoplasms , Calcium-Binding Proteins , Carcinoma, Non-Small-Cell Lung , Cathepsin F , Lung Neoplasms , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Brain Neoplasms/blood , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Calcium-Binding Proteins/blood , Calcium-Binding Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cathepsin F/blood , Cathepsin F/metabolism , Humans , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Prognosis , Up-Regulation
7.
Clin Epigenetics ; 13(1): 220, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34906185

ABSTRACT

BACKGROUND: Early lung cancer detection remains a clinical challenge for standard diagnostic biopsies due to insufficient tumor morphological evidence. As epigenetic alterations precede morphological changes, expression alterations of certain imprinted genes could serve as actionable diagnostic biomarkers for malignant lung lesions. RESULTS: Using the previously established quantitative chromogenic imprinted gene in situ hybridization (QCIGISH) method, elevated aberrant allelic expression of imprinted genes GNAS, GRB10, SNRPN and HM13 was observed in lung cancers over benign lesions and normal controls, which were pathologically confirmed among histologically stained normal, paracancerous and malignant tissue sections. Based on the differential imprinting signatures, a diagnostic grading model was built on 246 formalin-fixed and paraffin-embedded (FFPE) surgically resected lung tissue specimens, tested against 30 lung cytology and small biopsy specimens, and blindly validated in an independent cohort of 155 patients. The QCIGISH diagnostic model demonstrated 99.1% sensitivity (95% CI 97.5-100.0%) and 92.1% specificity (95% CI 83.5-100.0%) in the blinded validation set. Of particular importance, QCIGISH achieved 97.1% sensitivity (95% CI 91.6-100.0%) for carcinoma in situ to stage IB cancers with 100% sensitivity and 91.7% specificity (95% CI 76.0-100.0%) noted for pulmonary nodules with diameters ≤ 2 cm. CONCLUSIONS: Our findings demonstrated the diagnostic value of epigenetic imprinting alterations as highly accurate translational biomarkers for a more definitive diagnosis of suspicious lung lesions.


Subject(s)
Biomarkers, Tumor/genetics , Lung Neoplasms/diagnosis , Multiple Pulmonary Nodules/genetics , Aged , Biomarkers, Tumor/analysis , DNA Methylation/genetics , Early Detection of Cancer/methods , Early Detection of Cancer/standards , Early Detection of Cancer/statistics & numerical data , Epigenesis, Genetic/genetics , Female , Genomic Imprinting/genetics , Genomic Imprinting/physiology , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Multiple Pulmonary Nodules/etiology
8.
Clin Transl Med ; 11(9): e517, 2021 09.
Article in English | MEDLINE | ID: mdl-34586745

ABSTRACT

BACKGROUND: Platinum-based chemotherapy is effective in inducing shrinkage of primary lung cancer lesions; however, it shows finite therapeutic efficacy in patients suffering from brain metastasis (BM). The intrinsic changes of BM cells, which contribute to the poor results remain unknown. METHODS: Platinum drug-sensitivity was assessed by utilizing a preclinical BM model of PC9 lung adenocarcinoma cells in vitro and in vivo. High consumption of glutathione (GSH) and two associated upregulated proteins (GPX4 and GSTM1) in BM were identified by integrated metabolomics and proteomics in cell lines and verified by clinical serum sample. Gain-of-function and rescue experiments were implemented to reveal the impact and mechanism of GPX4 and GSTM1 on the chemosensitivity in BM. The interaction between GPX4 and GSTM1 was examined by immunoblotting and immunoprecipitation. The mechanism of upregulation of GPX4 was further uncovered by luciferase reporter assay, immunoprecipitation, and electrophoretic mobility shift assay. RESULTS: The derivative brain metastatic subpopulations (PC9-BrMs) of parental cells PC9 developed obvious resistance to platinum. Radically altered profiles of BM metabolism and protein expression compared with primary lung cancer cells were described and GPX4 and GSTM1 were identified as being responsible for the high consumption of GSH, leading to decreased chemosensitivity by negatively regulating ferroptosis. Besides, GSTM1 was found regulated by GPX4, which was transcriptionally activated by the Wnt/NR2F2 signaling axis in BM. CONCLUSIONS: Collectively, our findings demonstrated that Wnt/NR2F2/GPX4 promoted acquired chemoresistance by suppressing ferroptosis with high consumption of GSH. GPX4 inhibitor was found to augment the anticancer effect of platinum drugs in lung cancer BM, providing novel strategies for lung cancer patients with BM.


Subject(s)
Brain Neoplasms , Drug Resistance, Neoplasm , Lung Neoplasms , Phospholipid Hydroperoxide Glutathione Peroxidase , Platinum/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Ferroptosis/genetics , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
9.
Cell Death Dis ; 12(2): 214, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637678

ABSTRACT

Cancer-associated fibroblasts (CAFs) contribute to tumour epithelial-mesenchymal transition (EMT) via interaction with cancer cells. However, the molecular mechanisms underlying tumour-promoting EMT of CAFs in lung adenocarcinoma (ADC) remain unclear. Here, we observed that CAFs isolated from lung ADC promoted EMT via production of stromal cell-derived factor-1 (SDF-1) in conditioned medium (CM). CAF-derived SDF-1 enhanced invasiveness and EMT by upregulating CXCR4, ß-catenin, and PPARδ, while downregulating these proteins reversed the effect. Furthermore, RNAi-mediated CXCR4 knockdown suppressed ß-catenin and PPARδ expression, while ß-catenin inhibition effectively downregulated PPARδ without affecting CXCR4; however, treatment with a PPARδ inhibitor did not inhibit CXCR4 or ß-catenin expression. Additionally, pairwise analysis revealed that high expression of CXCR4, ß-catenin, and PPARδ correlated positively with 75 human lung adenocarcinoma tissues, which was predictive of poor prognosis. Thus, targeting the CAF-derived, SDF-1-mediated CXCR4 ß-catenin/ PPARδ cascade may serve as an effective targeted approach for lung cancer treatment.


Subject(s)
Adenocarcinoma of Lung/metabolism , Cancer-Associated Fibroblasts/metabolism , Chemokine CXCL12/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , PPAR delta/metabolism , Paracrine Communication , Receptors, CXCR4/metabolism , beta Catenin/metabolism , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adult , Aged , Aged, 80 and over , Cancer-Associated Fibroblasts/pathology , Coculture Techniques , Culture Media, Conditioned/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , PPAR delta/genetics , Prognosis , Receptors, CXCR4/genetics , Signal Transduction , Tumor Cells, Cultured , Up-Regulation , beta Catenin/genetics
10.
Micromachines (Basel) ; 12(1)2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33401770

ABSTRACT

Circulating tumor cells (CTCs) have been considered as an alternative to tissue biopsy for providing both germline-specific and tumor-derived genetic variations. Single-cell analysis of CTCs enables in-depth investigation of tumor heterogeneity and individualized clinical assessment. However, common CTC enrichment techniques generally have limitations of low throughput and cell damage. Herein, based on micropore-arrayed filtration membrane and microfluidic chip, we established an integrated CTC isolation platform with high-throughput, high-efficiency, and less cell damage. We observed a capture rate of around 85% and a purity of 60.4% by spiking tumor cells (PC-9) into healthy blood samples. Detection of CTCs from lung cancer patients demonstrated a positive detectable rate of 87.5%. Additionally, single CTCs, ctDNA and liver biopsy tissue of a representative advanced lung cancer patient were collected and sequenced, which revealed comprehensive genetic information of CTCs while reflected the differences in genetic profiles between different biological samples. This work provides a promising tool for CTCs isolation and further analysis at single-cell resolution with potential clinical value.

11.
Ann Transl Med ; 8(18): 1130, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33240979

ABSTRACT

BACKGROUND: Lung adenocarcinoma (LUAD) is still one of the major causes of cancer-related mortality across the globe. Therefore, there is a dire need to identify early specific and sensitive biomarkers or drug targets of LUAD for developing improved diagnosis and clinical management. We aimed to investigate the role of methyltransferase-like 7B (METTL7B) on LUAD tumor development and progression in this study. METHODS: METTL7B's expression was confirmed in two independent clinical cohort samples, including LUAD tissues microarray (TMA) via immunohistochemistry (IHC) and serum samples via enzyme-linked immunosorbent assay (ELISA). The correlation between METTL7B expression with clinicopathological features and overall survival rate in LUAD patients was then further analyzed. Meanwhile, the messenger ribonucleic acid (mRNA) and protein levels of METTL7B were verified in cell lines and in vitro experiments, including cell proliferation assay, and migration. Invasion assays were conducted to explore the effects of METTL7B on LUAD by silencing the protein expression. RESULTS: METTL7B was remarkably overexpressed in clinical LUAD tumor tissues and serum compared to the normal control group and in LUAD cell lines. The expression level of METTL7B was significantly correlated with tumor size, advanced tumor node and metastases (TNM) stages, and lymph node metastasis. The Kaplan-Meier survival curves proved that high METTL7B expression was significantly associated with a reduced survival rate in LUAD patients (P<0.05), and univariate analysis showed that high METTL7B expression was significantly associated with poor overall survival [hazard ratio (HR) =2.220, 95% confidence interval (CI): 1.211-4.086; P=0.010]. In vitro assays showed that METTL7B overexpression augmented cell proliferation, migration, and the invasion in LUAD. CONCLUSIONS: METTL7B was overexpressed in LUAD and significantly associated with the poor progression, showing that METTL7B may serve as a potential novel biomarker for the diagnosis and prognosis of LUAD. Moreover, METTL7B plays a role in promoting tumor proliferation, migration, and invasion in LUAD.

12.
Cytometry A ; 97(1): 46-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31595638

ABSTRACT

Circulating tumor cells (CTCs) represent invasive tumor cell populations and provide a noninvasive solution to the clinical management and research of tumors. Characterization of CTCs at single-cell resolution enables the comprehensive understanding of tumor heterogeneity and may benefit the diagnosis and treatment of cancer patients. However, most efforts have been made on enumeration and detection of CTCs, while little focus has been directed to single-cell study. Herein, an integrated microfluidic platform for single-cell isolation and analysis was established. After validating this platform on lung cancer cell lines, we detected and isolated single CTCs from the peripheral blood samples of 20 cancer patients before and after one treatment cycle. Furthermore, we performed single-cell whole-exome DNA sequencing on a single CTC from the peripheral blood sample of a representative early stage lung cancer patient. Among the blood samples of 20 patients, 15 of them were positive for CTC detection (75.0% detectable rate). Single-cell analysis revealed detailed genetic variations of the CTC, while six new gene mutations were detected in both single CTC and surgical specimen. This study provides a useful tool for the isolation and analysis of single CTCs from peripheral blood samples, which not only facilitates the early diagnosis of cancers but also helps to unravel the genetic information of tumor at a single-cell level. © 2019 International Society for Advancement of Cytometry.


Subject(s)
Lung Neoplasms/pathology , Microfluidics , Neoplastic Cells, Circulating/pathology , Single-Cell Analysis , Aged , Cell Line, Tumor , Cell Separation/methods , Female , Humans , Male , Microfluidic Analytical Techniques , Microfluidics/methods , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Single-Cell Analysis/methods
13.
Cell Death Dis ; 10(6): 377, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097690

ABSTRACT

The mechanism by which tumor-associated macrophages (TAMs) affect cancer progression is not fully understood. This study developed a microfluidic-based co-culture device to mimic the tumor microenvironment to assess TAM effects on invasion and metastasis in NSCLC. The results showed lung carcinoma cells could cause macrophages to show the M2 (a TAM-like) phenotype, and these M2 macrophages promoted lung cancer cell EMT and invasion. Proteomic analysis by the iTRAQ quantitation strategy and GO ontology of the cancer cells indicated that αB-Crystallin (CRYAB) might be involved in this process. Further, we confirmed the role of CRYAB in cancer invasion and metastasis through cell and animal experiments, as well as human cancer tissue assessment. Overall, we demonstrated that M2 macrophages promote malignancy in lung cancer through the EMT by upregulating CRYAB expression and activating the ERK1/2/Fra-1/slug signaling pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition , Macrophages/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Cell Line, Tumor , Coculture Techniques , Down-Regulation , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Microfluidics , Neoplasm Invasiveness , Neoplasm Metastasis , Proteomics , Signal Transduction , Tumor Microenvironment , Up-Regulation , alpha-Crystallin B Chain/genetics
14.
Cancer Med ; 7(2): 325-335, 2018 02.
Article in English | MEDLINE | ID: mdl-29356357

ABSTRACT

It is desirable to have a biomarker which can facilitate low-dose CT in diagnosis of early stage lung cancer. CTAPIII/CXCL7 is reported to be a potential biomarker for diagnosis of early lung cancer. In this study, we investigated the serum level of CTAPIII/CXCL7 in patients at different stage of lung cancer and the diagnostic efficacy of CTAPIII/CXCL7 in NSCLC. The plasma level of CTAPIII/CXCL7 was assayed by ELISA. CEA, SCCAg, and Cyfra211 were measured using a commercial chemiluminescent microparticle immunoassay. A total of 419 subjects were recruited, including 265 NSCLC patients and 154 healthy individuals. The subjects were randomly assigned to a training set and a test set. Receiver operating characteristic (ROC) and binary logistic regression analyses were conducted to evaluate the diagnostic efficacy and establish diagnostic mathematical model. Plasma CTAPIII/CXCL7 levels were significantly higher in NSCLC patients than in controls, which was independent of the stage of NSCLC. The diagnostic efficiency of CTAPIII/CXCL7 in NSCLC (training set: area under ROC curve (AUC) 0.806, 95% CI: 0.748-0.863; test set: AUC 0.773, 95% CI: 0.711-0.835) was greater than that of SCCAg, Cyfra21-1, or CEA. The model combining CTAPIII/CXCL7 with CEA, SCCAg, and Cyfra21-1 was more effective for NSCLC diagnosis than CTAPIII/CXCL7 alone. In addition, plasma level of CTAPIII/CXCL7 may contribute to the early diagnosis of NSCLC. CTAPIII/CXCL7 can be used as a plasma biomarker for the diagnosis of NSCLCs, particularly early stage lung cancer, with relatively high sensitivity and specificity.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Early Detection of Cancer/methods , Lung Neoplasms/diagnosis , beta-Thromboglobulin/analysis , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/blood , Case-Control Studies , Female , Follow-Up Studies , Humans , Lung Neoplasms/blood , Male , Middle Aged , Prognosis , ROC Curve
15.
Sci Rep ; 7: 42612, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198402

ABSTRACT

Recently there has been a more focus on the development of an efficient technique for detection of circulating tumor cells (CTCs), due to their significance in prognosis and therapy of metastatic cancer. However, it remains a challenge because of the low count of CTCs in the blood. Herein, a rapid and high-sensitivity approach for CTCs detection using an integrated microfluidic system, consisting of a deterministic lateral displacement (DLD) isolating structure, an automatic purifying device with CD45-labeled immunomagnetic beads and a capturing platform coated with rat-tail collagen was reported. We observed high capture rate of 90%, purity of about 50% and viability of more than 90% at the high throughput of 1 mL/min by capturing green fluorescent protein (GFP)-positive cells from blood. Further capturing of CTCs from metastatic cancers patients revealed a positive capture rate of 83.3%. Furthermore, our device was compared with CellSearch system via parallel analysis of 30 cancer patients, to find no significant difference between the capture efficiency of both methods. However, our device displayed advantage in terms of time, sample volume and cost for analysis. Thus, our integrated device with sterile environment and convenient use will be a promising platform for CTCs detection with potential clinical application.


Subject(s)
Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Neoplasms/diagnosis , Neoplastic Cells, Circulating/pathology , Aged , Aged, 80 and over , Biomarkers , Case-Control Studies , Cell Line, Tumor , Cell Survival , Combined Modality Therapy , Female , Fluorescent Antibody Technique , Humans , Male , Microscopy, Fluorescence , Middle Aged , Neoplasm Staging , Neoplasms/therapy
16.
ACS Appl Mater Interfaces ; 8(39): 25840-25847, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27606718

ABSTRACT

Metastasis is a complex pathophysiological process. As the main cause of cancer mortality in humans it represents a serious challenge to both basic researchers and clinicians. Here we report the design and construction of a multi-organ microfluidic chip that closely mimics the in vivo microenvironment of lung cancer metastasis. This multi-organs-on-a-chip includes an upstream "lung" and three downstream "distant organs", with three polydimethylsiloxane (PDMS) layers and two thin PDMS microporous membranes bonded to form three parallel microchannels. Bronchial epithelial, lung cancer, microvascular endothelial, mononuclear, and fibroblast cells were grown separated by the biomembrane in upstream "lung", while astrocytes, osteocytes, and hepatocytes were grown in distant chambers, to mimic lung cancer cell metastasis to the brain, bone, and liver. After culture in this system, lung cancer cells formed a "tumor mass", showed epithelial-mesenchymal transition (with altered expression of E-cadherin, N-cadherin, Snail1, and Snail2) and invasive capacity. A549 cells co-cultured with astrocytes overexpressed CXCR4 protein, indicating damage of astrocytes after cancer cell metastasis to the brain. Osteocytes overexpressed RANKL protein indicates damage of osteocytes after cancer cell metastasis to the bone, and hepatocytes overexpressed AFP protein indicates damage to hepatocytes after cancer cell metastasis to the liver. Finally, in vivo imaging of cancer growth and metastasis in a nude mice model validated the performance of metastasis in the organs-on-chip system. This system provides a useful tool to mimic the in vivo microenvironment of cancer metastasis and to investigate cell-cell interactions during metastasis.


Subject(s)
Microfluidics , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Lung , Lung Neoplasms , Mice , Mice, Nude , Neoplasm Metastasis , Tumor Microenvironment
17.
Sci Rep ; 6: 29719, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27411490

ABSTRACT

CD47 is overexpressed in many human cancers, its level positively correlates with tumor invasion and metastasis. However, it is largely unknown whether CD47 overexpression drives metastasis and how CD47 lead to tumor metastasis in non-small cell lung cancer (NSCLC). In this study, we analyzed NSCLC specimens and cell lines, and revealed that CD47 is expressed at a higher level than in tumor-free control samples. Furthermore, increased CD47 expression correlated with clinical staging, lymph node metastasis and distant metastasis. In order to understand the molecular mechanisms underlying CD47 functions, we applied both gain-of-function and loss-of-function approaches in cell lines. The siRNA-mediated downregulation of CD47 inhibited cell invasion and metastasis in vitro, while the overexpression of CD47 by plasmid transfection generated opposite effects. In vivo, CD47-specific shRNA significantly reduced tumor growth and metastasis. On the molecular level, the expression of CD47 correlated with that of Cdc42, both in cell lines and NSCLC specimens. The inhibition of Cdc42 attenuates the invasion and metastasis of CD47-overexpressing cells. These results indicate that Cdc42 is a downstream mediator of CD47-promoted metastasis. Our findings provide first evidence that CD47 is an adverse prognostic factor for disease progression and metastasis, and a promising therapeutic target for NSCLC.


Subject(s)
CD47 Antigen/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , A549 Cells , Animals , CD47 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , RNA Interference , RNAi Therapeutics/methods , Xenograft Model Antitumor Assays/methods
18.
PLoS One ; 10(6): e0129593, 2015.
Article in English | MEDLINE | ID: mdl-26115510

ABSTRACT

Tumor stroma and growth factors provide a survival environment to tumor cells and can modulate their chemoresistance by dysregulating several signal pathways. In this study, we fabricated a three-dimensional (3D) microfluidic chip using polydimethylsiloxane (PDMS) to investigate the impact of hepatocyte growth factor (HGF) from cancer-associated fibroblasts (CAF) on the Met/PI3K/AKT activation, glucose regulatory protein (GRP78) expression and the paclitaxel-induced A549 cell apoptosis. With a concentration gradient generator, the assembled chip was able to reconstruct a tumor microenvironment in vitro. We found high levels of HGF in the supernatants of CAF and the CAF matrix from the supernatants of activated HFL1 fibroblasts or HGF enhanced the levels of Met, PI3K and AKT phosphorylation and GRP78 expression in A549 cells cultured in a 3D cell chamber, which was abrogated by anti-HGF. Inhibition of Met attenuated the CAF matrix-enhanced PI3K/AKT phosphorylation and GRP78 expression while inhibition of PI3K reduced GRP78 expression, but not Met phosphorylation in A549 cells. Inhibition of GRP78 failed to modulate the CAF matrix-enhanced Met/PI3K/AKT phosphorylation in A549 cells. Furthermore, inhibition of PI3K or GRP78 enhanced spontaneous and paclitaxel-induced A549 cell apoptosis. Moreover, treatment with the CAF matrix inhibited spontaneous and medium or high dose of paclitaxel-induced A549 cell apoptosis. Inhibition of PI3K or GRP78 attenuated the CAF matrix-mediated inhibition on paclitaxel-induced A549 cell apoptosis. Our data indicated that HGF in the CAF matrix activated the Met/PI3K/AKT and up-regulated GRP78 expression, promoting chemoresistance to paclitaxel-mediated apoptosis in A549 cells. Our findings suggest that the microfluidic system may represent an ideal platform for signaling research and drug screening.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factors/metabolism , Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Cell Culture Techniques , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Fibroblast Growth Factors/pharmacology , Fibroblasts/metabolism , Humans , Microfluidics/methods , Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Oncotarget ; 6(11): 8900-13, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25823926

ABSTRACT

We investigated the role of macrophages in promoting benzopyrene (BaP)-induced malignant transformation of human bronchial epithelial cells using a BaP-induced tumor transformation model with a bionic airway chip in vitro and in animal models. The bionic airway chip culture data showed that macrophages promoted BaP-induced malignant transformation of human bronchial epithelial cells, which was mediated by nuclear factor (NF)-κB and STAT3 pathways to induce cell proliferation, colony formation in chip culture, and tumorigenicity in nude mice. Blockage of interleukin (IL)-6 or tumor necrosis factor (TNF)-α signaling or inhibition of NF-κB, STAT3, or cyclinD1 expression abrogated the effect of macrophages on malignant transformation in the bionic airway chip culture. In vivo, macrophages promoted lung tumorigenesis in a carcinogen-induced animal model. Similarly, blockage of NF-κB, STAT3, or cyclinD1 using siRNA transfection decreased the carcinogen-induced tumorigenesis in rats. We demonstrated that macrophages are critical in promoting lung tumorigenesis and that the macrophage-initiated TNF-α/NF-κB/cyclinD1 and IL-6/STAT3/cyclinD1 pathways are primarily responsible for promoting lung tumorigenesis.


Subject(s)
Adenocarcinoma/pathology , Bronchi/drug effects , Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic/drug effects , Lab-On-A-Chip Devices , Lung Neoplasms/pathology , Macrophages/physiology , NF-kappa B/physiology , Neoplasm Proteins/physiology , STAT3 Transcription Factor/physiology , Aged , Animals , Benzo(a)pyrene , Bronchi/pathology , Cell Count , Cell Transformation, Neoplastic/genetics , Cyclin D1/antagonists & inhibitors , Cyclin D1/physiology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/physiology , Lung Neoplasms/chemically induced , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Rats , Rats, Wistar , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Tobacco Smoke Pollution/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL