Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nat Commun ; 15(1): 1200, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331898

RESUMEN

The complexity of the tumor microenvironment (TME) is a crucial factor in lung adenocarcinoma (LUAD) progression. To gain deeper insights into molecular mechanisms of LUAD, we perform an integrative single-cell RNA sequencing (scRNA-seq) data analysis of 377,574 cells from 117 LUAD patient samples. By linking scRNA-seq data with bulk gene expression data, we identify a cluster of prognostic-related UPP1high tumor cells. These cells, primarily situated at the invasive front of tumors, display a stronger association with the immunosuppressive components in the TME. Our cytokine array analysis reveals that the upregulation of UPP1 in tumor cells leads to the increased release of various immunosuppressive cytokines, with TGF-ß1 being particularly prominent. Furthermore, this UPP1 upregulation also elevates the expression of PD-L1 through the PI3K/AKT/mTOR pathway, which contributes to the suppression of CD8 + T cells. Cytometry by time-of-flight (CyTOF) analysis provides additional evidence of the role of UPP1 in shaping the immunosuppressive nature of the TME. Using patient-derived organoids (PDOs), we discover that UPP1high tumors exhibit relatively increased sensitivity to Bosutinib and Dasatinib. Collectively, our study highlights the immunosuppressive role of UPP1 in LUAD, and these findings may provide insights into the molecular features of LUAD and facilitate the development of personalized treatment strategies.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Adenocarcinoma del Pulmón/genética , Linfocitos T CD8-positivos , Citocinas , Inmunosupresores , Neoplasias Pulmonares/genética , Fosfatidilinositol 3-Quinasas/genética , Microambiente Tumoral/genética
2.
Biochem Genet ; 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38206423

RESUMEN

The toll-like receptor (TLR) family is an important class of proteins involved in the immune response. However, little is known about the association between TLRs and Esophageal squamous cell cancer (ESCC). We explored differentially expressed genes (DEGs) between ESCC and esophagus tissues in TCGA and GTEx database. By taking the intersection with TLR gene set and using univariate Cox analysis and multivariate Cox regression analysis to discriminate the hub genes, we created a TLR-prognostic model. Our model separated patients with ESCC into high- and low-risk score (RS) groups. Prognostic analysis was performed with Kaplan-Meier curves. The two groups were also compared regarding tumor immune microenvironment and drug sensitivity. Six hub genes (including CD36, LGR4, MAP2K3, NINJ1, PIK3R1, and TRAF3) were screened to construct a TLR-prognostic model. High-RS group had a worse survival (p < 0.01), lower immune checkpoint expression (p < 0.05), immune cell abundance (p < 0.05) and decreased sensitivity to Epirubicin (p < 0.001), 5-fluorouracil (p < 0.0001), Sorafenib (p < 0.01) and Oxaliplatin (p < 0.05). We constructed a TLR-based model, which could be used to assess the prognosis of patients with ESCC, provide new insights into drug treatment for ESCC patients and investigate the TME and drug response.

3.
EBioMedicine ; 100: 104971, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38244291

RESUMEN

BACKGROUND: Neoadjuvant chemoimmunotherapy has offered novel therapeutic options for patients with locally advanced oesophageal squamous cell carcinoma (ESCC). Depicting the landscape of genomic and immune profiles is critical in predicting therapeutic responses. METHODS: We integrated whole-exome sequencing, single-cell RNA sequencing, and immunofluorescence data of ESCC samples from 24 patients who received neoadjuvant treatment with PD-1 inhibitors plus paclitaxel and platinum-based chemotherapy to identify correlations with therapeutic responses. FINDINGS: An elevation of small insertions and deletions was observed in responders. DNA mismatch repair (MMR) pathway alternations were highly frequent in patients with optimal responses and correlated with tumour infiltrating lymphocytes (TILs). Among the TILs in ESCC, dichotomous developing trajectories of B cells were identified, with one lineage differentiating towards LMO2+ germinal centre B cells and another lineage differentiating towards CD55+ memory B cells. While LMO2+ germinal centre B cells were enriched in responding tumours, CD55+ memory B cells were found to correlate with inferior responses to combination therapy, exhibiting immune-regulating features and impeding the cytotoxicity of CD8+ T cells. The comprehensive evaluation of transcriptomic B cell lineage features was validated to predict responses to immunotherapy in patients with cancer. INTERPRETATION: This comprehensive evaluation of tumour MMR pathway alternations and intra-tumoural B cell features will help to improve the selection and management of patients with ESCC to receive neoadjuvant chemoimmunotherapy. FUNDING: National Science Foundation of China (82373371, 82330053), Eastern Scholar Program at Shanghai Institutions of Higher Learning, National Science and Technology Major Project of China (2023YFA1800204, 2020YFC2008402), and Science and Technology Commission of Shanghai Municipality (22ZR1410700, 20ZR1410800).


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/genética , Terapia Neoadyuvante , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Linfocitos T CD8-positivos , Linaje de la Célula/genética , China , Genómica
4.
Commun Biol ; 6(1): 570, 2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37248295

RESUMEN

Ferroptosis, an iron-dependent non-apoptotic cell death, has been shown to play a vital role in tumor proliferation and chemotherapy resistance. Here, we report that KLF11 inhibits lung adenocarcinoma (LUAD) cell proliferation and promotes chemotherapy sensitivity by participating in the GPX4-related ferroptosis pathway. Through an RNA-sequence screen from LUAD cells pretreatment with ferroptosis inducers (FINs), we discovered that KLF11 expression was significantly higher in FINs-treated cells, suggesting that KLF11 may be involved in ferroptosis. Overexpression of KLF11 promoted LUAD cells to undergo ferroptosis alterations. Meanwhile, upregulation of KLF11 expression also inhibited cell proliferation and increased chemosensitivity, whereas knockout of KLF11 did the opposite. With ChIP-Seq and RNA-Seq, we identified GPX4 as a downstream target of KLF11. Through ChIP-qPCR and dual luciferase assay, we clarified that KLF11 binds to the promoter region of GPX4 and represses its transcription. Restored GPX4 expression antagonized the ability of KLF11 to promote ferroptosis, increase chemotherapy sensitivity and inhibit cell proliferation in vitro and in vivo. Clinically, KLF11 declined in LUAD and its low expression was associated with reduced patient survival. Our findings established the function of KLF11 to promote ferroptosis in LUAD, thereby inhibiting cell proliferation and enhancing the efficacy of chemotherapy.


Asunto(s)
Adenocarcinoma del Pulmón , Proteínas Reguladoras de la Apoptosis , Ferroptosis , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Proteínas Represoras , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Humanos , Línea Celular Tumoral , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Proliferación Celular , Mitocondrias
5.
Transl Lung Cancer Res ; 12(1): 141-149, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36762056

RESUMEN

Background: Pembrolizumab has been shown to be effective and safe in improving the survival of patients with advanced non-small-cell lung cancer (NSCLC). However, the effectiveness and safty of pembrolizumab in the induction treatment of patients with potential resectable clinical stage III NSCLC remains undetermined. Methods: A total of 25 patients who received neoadjuvant pembrolizumab plus chemotherapy for preoperative stage III NSCLC between August 2020 and November 2021 in Zhongshan Hospital were retrospectively evaluated, and 21 of them were followed by pulmonary resection. The neoadjuvant treatment was as follows: intravenous pembrolizumab (200 mg) on day 1, carboplatin [target area under the curve (AUC) 5 mg/mL] or cisplatin (75 mg/m2) on day 1, and pemetrexed (500 mg/m2 for adenocarcinoma) or nab-paclitaxel (260 mg/m2 for other subtypes) on day 1 of every 21-day cycle up to two or three cycles. Results: The mean age of all 25 patients was 65 years, of whom 22 were men and 3 were women. Seventeen were diagnosed before treatment as clinical stage IIIA, seven as IIIB, and one as IIB. All received neoadjuvant immunotherapy plus chemotherapy. Following induction therapy, 21 patients with stable disease or partial response (PR) according to the Response Evaluation Criteria in Solid Tumors (RECIST 1.1) underwent surgical resection without delay. Among the patients who underwent operation, major pathological response (MPR) was achieved in 13 patients, including 6 (28.6%) patients achieved a complete pathological response (CPR). Two patients with partial radiologic remission refused operative treatment, one had progressive disease (PD), and another developed a grade immune pneumonia and could not tolerate surgery. However, none of the adverse events caused surgery delays or deaths. Conclusions: Neoadjuvant pembrolizumab plus chemotherapy could be considered reliable for clinical stage III NSCLC, but needs to be validated with more robust clinical trials.

6.
Front Pharmacol ; 13: 1072589, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36467089

RESUMEN

Objective: The goal of this study was to create a risk model based on the ferroptosis gene set that affects lung adenocarcinoma (LUAD) patients' prognosis and to investigate the potential underlying mechanisms. Material and Methods: A cohort of 482 LUAD patients from the TCGA database was used to develop the prognostic model. We picked the module genes from the ferroptosis gene set using weighted genes co-expression network analysis (WGCNA). The least absolute shrinkage and selection operator (LASSO) and univariate cox regression were used to screen the hub genes. Finally, the multivariate Cox analysis constructed a risk prediction score model. Three other cohorts of LUAD patients from the GEO database were included to validate the prediction ability of our model. Furthermore, the differentially expressed genes (DEG), immune infiltration, and drug sensitivity were analyzed. Results: An eight-gene-based prognostic model, including PIR, PEBP1, PPP1R13L, CA9, GLS2, DECR1, OTUB1, and YWHAE, was built. The patients from the TCGA database were classified into the high-RS and low-RS groups. The high-RS group was characterized by poor overall survival (OS) and less immune infiltration. Based on clinical traits, we separated the patients into various subgroups, and RS had remarkable prediction performance in each subgroup. The RS distribution analysis demonstrated that the RS was significantly associated with the stage of the LUAD patients. According to the study of immune cell infiltration in both groups, patients in the high-RS group had a lower abundance of immune cells, and less infiltration was associated with worse survival. Besides, we discovered that the high-RS group might not respond well to immune checkpoint inhibitors when we analyzed the gene expression of immune checkpoints. However, drug sensitivity analysis suggested that high-RS groups were more sensitive to common LUAD agents such as Afatinib, Erlotinib, Gefitinib, and Osimertinib. Conclusion: We constructed a novel and reliable ferroptosis-related model for LUAD patients, which was associated with prognosis, immune cell infiltration, and drug sensitivity, aiming to shed new light on the cancer biology and precision medicine.

7.
J Cancer ; 13(10): 3140-3150, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36046648

RESUMEN

Background: Lung adenocarcinoma (LUAD) is the most prevalent thoracic cancer with the highest incidence and mortality worldwide. Baculoviral IAP Repeat Containing 5 (BIRC5) is well studied in many malignancies, its prognosis value and correlation with the tumor microenvironment (TME) in LAUD remains largely elusive. Methods: The Wilcoxon signed-rank test and logistic regression were used to evaluate the relationship between clinical features and BIRC5 expression in LUAD. To assess the impact of BIRC5 on prognosis, the Kaplan-Meier plotter analysis and Cox regression were used, as well as a receiver operating characteristic (ROC) curve and nomogram. Gene set enrichment analysis (GSEA) and single-sample gene set enrichment analysis (ssGSEA) were recruited to predict the association between BIRC5 and immune cell infiltrations. Furthermore, qRT-PCR and western bolt were utilized to confirm gene expression on mRNA and protein levels. The proliferation of A549 and H1299 cells was evaluated using CCK8 and EdU assay. Cell mobility was tested by transwell assay and wound healing assay. Detection of PD-L1 and infiltrated CD8 T cells in xenograft tumors was done by flow cytometry. Results: BIRC5 expression was found to be substantially greater in LUAD patients. According to KM-plotter analysis, patients with high levels of BIRC5 had shorter survival rates. Multivariate Cox analysis revealed that elevated BIRC5 expression was an independent risk factor for OS and PFS in LUAD patients. High BIRC5 expression was predicted to be associated with chemokine activity and immune cell chemotaxis, whereas ssGSEA suggested that BIRC5 is highly associated with CD8 T cell infiltration and PD-L1 levels. In vitro experiments suggested overexpression of BIRC5 promoted the proliferation, mobility, and PD-L1 level of A549 cells, and vice versa in H1299 cells. Furthermore, in vivo study suggested elevated tumor weight and PD-L1 levels in xenograft tumors generated from LLC cells with overexpressed BIRC5. Conclusion: BIRC5 promotes lung adenocarcinoma progression by modulating PD-L1 expression and inducing tumor immune evasion.

8.
BMC Pulm Med ; 22(1): 209, 2022 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-35619108

RESUMEN

BACKGROUND: Tumor spread through air spaces (STAS) has been shown to adversely affect the prognosis of lung cancer. The correlation between clinicopathological and genetic features and STAS remains unclear. METHOD: We retrospectively reviewed 3075 NSCLC patients between2017-2019. We evaluated the relationship between STAS and patients' clinicopathological and molecular features. The chi-square test was performed to compare categorical variables. Univariate analysis and multivariate logistic regression analysis were performed to investigate the association of clinical factors with STAS. A nomogram was formulated to predict the presence of STAS. RESULTS: STAS was identified in 617 of 3075 patients (20.07%). STAS was significantly related to sex (p < 0.001), smoking (p < 0.001), CEA (p < 0.001), differentiation (p < 0.001), histopathological type (p < 0.001), lymphatic vessel invasion (p < 0.001), pleural invasion (p < 0.001), T stage (p < 0.001), N stage (p < 0.001), M stage (p < 0.001), and TNM stage (p < 0.001). STAS was frequently found in tumors with wild-type EGFR (p < 0.001), KRAS mutations (p < 0.001), ALK rearrangements (p < 0.001) or ROS1 rearrangements (p < 0.001). For programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1), STAS was associated with PD-L1 expression level in tumor cells (p < 0.001) or stromal cells (p < 0.001), while PD-1 only in stromal cells (p < 0.001). Multivariable analyses demonstrated significant correlations between STAS and CEA level (p < 0.001), pathological grade (p < 0.001), lymphatic vessel invasion (p < 0.001), pleural invasion (p = 0.001), and TNM stage (p = 0.002). A nomogram was formulated based on the results of the multivariable analysis. CONCLUSIONS: Tumor STAS was associated with several invasive clinicopathological features. A nomogram was established to predict the presence of STAS in patients with NSCLC.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/patología , Antígeno B7-H1 , Antígeno Carcinoembrionario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Invasividad Neoplásica , Estadificación de Neoplasias , Nomogramas , Receptor de Muerte Celular Programada 1 , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas , Estudios Retrospectivos
9.
Front Microbiol ; 13: 859352, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35586863

RESUMEN

To figure out the molecular mechanism in the esophageal squamous carcinoma (ESCC) with the discrepancy in the tissue-resident microbiota, we selected clinical features, RNA sequences, and transcriptomes of ESCC patients from The Cancer Genome Atlas (TCGA) website and detailed tissue-resident microbiota information from The Cancer Microbiome Atlas (n = 60) and explored the infiltration condition of particular microbiota in each sample. We classified the tissue-resident micro-environment of ESCC into two clusters (A and B) and built a predictive classifier model. Cluster A has a higher proportion of certain tissue-resident microbiota with comparatively better survival, while Cluster B has a lower proportion of certain tissue-resident microbiota with comparatively worse survival. We showed traits of gene and clinicopathology in the esophageal tissue-resident micro-environment (ETM) phenotypes. By comparing the two clusters' molecular signatures, we find that the two clusters have obvious differences in gene expression and mutation, which lead to pathway expression discrepancy. Several pathways are closely related to tumorigenesis. Our results may demonstrate a synthesis of the infiltration pattern of the esophageal tissue-resident micro-environment in ESCC. We reveal the mechanism of esophageal tissue-resident microbiota discrepancy in ESCC, which may contribute to therapy progress for patients with ESCC.

10.
Cancer Immunol Immunother ; 71(10): 2511-2522, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35278107

RESUMEN

BACKGROUND: Immune checkpoint blockade (ICB) has been improving patient outcomes of non-small cell lung cancer (NSCLC), but its effectiveness is highly subjective to individual tumor microenvironment. As dominant immune cells in NSCLC, tumor-associated macrophages (TAMs) display high diversity and plasticity. This study aims to find crucial TAM subtypes associated with ICB response in NSCLC. MATERIALS AND METHODS: Large cohorts of NSCLC patients from The Cancer Genome Atlas and a single-cell sequencing dataset were integrated to illustrate immunosuppressive phenotypes of TAMs, followed by experimental verification. 341 NSCLC surgical samples and 40 tissue samples of NSCLC patients who received ICB treatment were collected to state the clinical importance of TAMs. RESULTS: We identified a TREM2 positive (+) TAM subtype in NSCLC stratifying patient responses to immunotherapy. NSCLC patients with high TREM2+ TAM infiltration exhibited advanced tumor progression, inferior prognosis and unique NSCLC molecular characteristics, especially mutations of EGFR. TREM2+ TAMs were induced in TME, but not existed in peripheral blood. TREM2+ TAMs were enriched with multiple anti-inflammatory cytokines, exhibiting a M2-like immunosuppressive phenotype, and potentiate T cell dysfunction including impaired anti-tumor activity of CD8+ T cells and enhanced differentiation towards FOXP3+ Tregs, thus facilitating immune evasion of NSCLC. Response rates to PD-1-based ICB were higher in patients with low TREM2+ TAMs (31.58%) compared to high TREM2+ TAMs (14.29%). CONCLUSIONS: Our findings implicated the immunosuppressive role of TREM2+ TAMs in NSCLC, and systematically reveal the clinical significance of TREM2+ TAMs as predictive and prognostic markers for NSCLC patients with ICB treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Linfocitos T CD8-positivos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Factores Inmunológicos , Inmunoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Macrófagos , Glicoproteínas de Membrana , Pronóstico , Receptores Inmunológicos , Microambiente Tumoral
11.
EBioMedicine ; 70: 103510, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34365093

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) remains one of the deadly cancer types. Comprehensively dissecting the molecular characterization and the heterogeneity of ESCC paves the way for developing more promising therapeutics. METHODS: Expression profiles of multiple ESCC datasets were integrated. ATAC-seq and RNA-seq were combined to reveal the chromatin accessibility features. A prognosis-related subtype classifier (PrSC) was constructed, and its association with the tumor microenvironment (TME) and immunotherapy was assessed. The key gene signature was validated in clinical samples. Based on the TME heterogeneity of ESCC patients, potential subtype-specific therapeutic agents were screened. FINDINGS: The common differentially expressed genes (cDEGs) in ESCC were identified. Up-regulated genes (HEATR1, TIMELESS, DTL, GINS1, RUVBL1, and ECT2) were found highly important in ESCC cell survival. The expression alterations of PRIM2, HPGD, NELL2, and TFAP2B were associated with chromatin accessibility changes. PrSC was a robust scoring tool that was not only associated with the prognosis of ESCC patients, but also could reflect the TME heterogeneity. TNS1high fibroblasts were associated with immune exclusion. TG-101348 and Vinorelbine were identified as potential subtype-specific therapeutic agents. Besides, the application of PrSC into two immunotherapy cohorts indicated its potential value in assessing treatment response to immunotherapy. INTERPRETATION: Our study depicted the multi-dimensional characterization of ESCC, established a robust scoring tool for the prognosis assessment, highlighted the role of TNS1high fibroblasts in TME, and identified potential drugs for clinical use. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma/genética , Neoplasias Esofágicas/genética , Farmacología en Red/métodos , Transcriptoma , Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Microambiente Tumoral/efectos de los fármacos , Vinorelbina/farmacología
12.
Brief Bioinform ; 22(4)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33003204

RESUMEN

N6-methyladenosine (m6A) modification can regulate a variety of biological processes. However, the implications of m6A modification in lung adenocarcinoma (LUAD) remain largely unknown. Here, we systematically evaluated the m6A modification features in more than 2400 LUAD samples by analyzing the multi-omics features of 23 m6A regulators. We depicted the genetic variation features of m6A regulators, and found mutations of FTO and YTHDF3 were linked to worse overall survival. Many m6A regulators were aberrantly expressed in tumors, among which FTO, IGF2BP3, YTHDF1 and RBM15 showed consistent alteration features across 11 independent cohorts. Besides, the regulator-pathway interaction network demonstrated that m6A modification was associated with various biological pathways, including immune-related pathways. The correlation between m6A regulators and tumor microenvironment was also assessed. We found that LRPPRC was negatively correlated with most tumor-infiltrating immune cells. On the other hand, we established a scoring tool named m6Sig, which was positively correlated with PD-L1 expression and could reflect both the tumor microenvironment characterization and prognosis of LUAD patients. Comparison of CNV between high and low m6Sig groups revealed differences on chromosome 7. Application of m6Sig on an anti-PD-L1 immunotherapy cohort confirmed that the high m6Sig group demonstrated therapeutic advantages and clinical benefits. Our study indicated that m6A modification is involved in many aspects of LUAD and contributes to tumor microenvironment formation. A better understanding of m6A modification will provide more insights into the molecular mechanisms of LUAD and facilitate developing more effective personalized treatment strategies. A web application was built along with this study (http://www.bioinfo-zs.com/luadexpress/).


Asunto(s)
Adenina/análogos & derivados , Adenocarcinoma del Pulmón , Bases de Datos de Ácidos Nucleicos , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Proteínas de Neoplasias , Adenina/metabolismo , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética
13.
Ann Palliat Med ; 10(2): 1560-1568, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33183042

RESUMEN

BACKGROUND: Anastomotic leakage with empyema is one of the most severe complications of esophagectomy. We invented a new type of slow-flow irrigation drainage tube to improve the smooth pleural drainage, which contributed to the recovery of anastomotic leakage. METHODS: In this study, 42 patients, from 2012 to 2019, who underwent esophagectomy and postoperative anastomotic leakage with persistent empyema, were enrolled and distributed into irrigation drainage tube group (I+) or non-irrigation drainage tube group (I-). In (I+) group, the slow-flow irrigation drainage tube was placed along the primary pleural drainage tube into the empyema cavity, irrigated with normal saline. Clinicopathological features and perioperative complications of all patients were collected and compared to determine the differences between the two groups with statistical analysis. RESULTS: All 20 patients were distributed into (I+) group and 22 into (I-) group. The two groups were comparable in clinicopathological features. When compared with (I-) group, patients in (I+) group achieved significantly shorter healing duration and postoperative hospital stay. The complications resulting from anastomotic leakage in (I+) group were relatively fewer than those in (I-) group, including postoperative bleeding (0 vs. 1) and pulmonary complications (7 vs. 13). Moreover, 5 reoperations for rescuing bleeding or severe thoracic infection were performed in (I-) group, while none in (I+) group. CONCLUSIONS: The slow-flow irrigation drainage tube is a safe and effective treatment of anastomotic leakage and empyema. Additional prospective study is required to furtherly determine the improvement of drainage and extended application in deep infections located in thorax.


Asunto(s)
Empiema , Neoplasias , Fuga Anastomótica/etiología , Fuga Anastomótica/terapia , Drenaje , Empiema/etiología , Empiema/terapia , Unión Esofagogástrica/cirugía , Humanos , Complicaciones Posoperatorias/terapia , Estudios Prospectivos , Estudios Retrospectivos
14.
Cell Commun Signal ; 18(1): 17, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32014008

RESUMEN

BACKGROUND: Esophageal squamous cell cancer (ESCC) is one kind of frequent digestive tumor. The inflammatory environment plays an important role in the tumorigenesis and development of ESCC. Cancer stem cells are a small group of tumor cells with stem cell characteristics, which can potentially hinder the tumor management and treatment. METHODS: ELISA was performed to detect the lipopolysaccharide concentration in cancer tissues. qPCR, Western blot, FACS, Immunohistochemistry, Immunofluorescence and Dot blot were applied to detect target genes expression. CCK-8, Colony-formation, Transwell, Sphere and Xenograft were conducted to investigate the function of cells, influenced by risk factors. The survival curve was drawn with the Kaplan-Meier product limit estimator. Nano-hmC-Seal-seq was utilized to detect the downstream target of TET3. ChIP-qPCR was adopted to demonstrate the transcriptional regulation of stem cell-associated genes by HOXB2. RESULTS: Lipopolysaccharide concentration was significantly up-regulated in ESCC. High concentration of lipopolysaccharide stimulation induced the stemness of ESCC cells. TET3 expression was elevated with lipopolysaccharide stimulation via p38/ERK-MAPK pathway in ESCC and negatively correlated with patients' survival. TET3 induced the stemness of ESCC cells. Nano-hmC-Seal-seq showed that TET3 overexpression led to a significant increase in 5hmC levels of HOXB2 gene region, which was thus identified as the downstream target of TET3. The binding of HOXB2 to NANOG and cMYC was verified by ChIP-qPCR. CONCLUSIONS: Lipopolysaccharide served as a tumor promotor in ESCC by inducing cancer cell stemness through the activation of a LPS-TET3-HOXB2 signaling axis, which might provide a novel therapeutic strategy for ESCC. Video Abstract.


Asunto(s)
Dioxigenasas/metabolismo , Epigénesis Genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Proteínas de Homeodominio/metabolismo , Lipopolisacáridos/farmacología , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Desnudos , Persona de Mediana Edad , Análisis Multivariante , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Transcripción Genética/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Cancer ; 11(4): 810-818, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31949484

RESUMEN

Background: Primary adenosquamous carcinoma (ASC) is a rare malignant tumor in the lung and its biological behavior has not yet been thoroughly described. In this study, we aimed to explore the clinical and biological role of CXCR4 in patients with resected lung ASC. Methods: We retrospectively reviewed the clinical records of patients with histologically confirmed lung ASC who underwent surgical resection with systematic lymph node dissection. Immunohistochemical staining was performed to detect the expression of CXCR4 in tumor tissues. The correlation between CXCR4 expression and clinicopathological characteristics were evaluated. The association between CXCR4 expression and patients' prognosis was analyzed by Kaplan-Meier and Cox regression. Moreover, we performed in vitro studies including CCK8, transwell and cell apoptosis to explore the potential role of CXCR4 in lung ASC. Results: A total of 78 patients with resected lung ASC were reviewed. Seventy (89.7%) patient tumors expressed CXCR4, with high level of CXCR4 expression observed in 45 (57.7%) cases. In vitro, CXCR4 conferred no difference in proliferative capacity but increased invasive potential, enhanced chemoresistance and inhibited apoptosis of lung ASC. Clinically, high CXCR4 expression was significantly associated with solid ASC, lymph node metastasis and advanced TNM stage. Patients with high CXCR4 expression and solid ASC had decreased disease-free survival and overall survival.Conclusions: CXCR4 was commonly expressed in lung ASC tumors. High CXCR4 expression might be a novel marker in predicting a poor prognosis in resected lung ASC and might serve as a potential therapeutic target.

16.
BMC Cancer ; 19(1): 875, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31481019

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is one of the most frequent malignant digestive tumors around the world. We previously demonstrated that eIF3b could promote the progression of ESCC. The exact mechanisms underlying these effects remained unknown. METHODS: Quantitative proteomics was applied to detect the potential targets of Eukaryotic translation initiation factor 3 subunit b (eIF3b). RT-qPCR and Western blot were performed to detect the expression of targeted gene and pathway related genes. RNA-immunoprecipitation was applied to verify the binding of eIF3b with targeted gene. Moreover, CCK-8 assay, colony-formation assay, transwell assay, flow cytometry for cell apoptosis and tumor xenograft assay were performed to analyze the regulation of the targeted gene on the bio-function of ESCC cells. RESULTS: Quantitative proteomics data showed that Testis-expressed protein 9 (TEX9) expression was positively associated with eIF3b expression. RT-qPCR and Western blot results confirmed the quantitative proteomics data and demonstrated that TEX9 expression was positively correlated with TNM stage in ESCC. Furtherly, RNA-immunoprecipitation confirmed that eIF3b binding to TEX9 mRNA. The bio-function related assay demonstrated that TEX9 and eIF3b functionally synergized to promote the proliferation and migration, and inhibited the apoptosis of ESCC cells. In the analysis of mechanism, we revealed that TEX9 and eIF3b promoted the progression of ESCC through the activation of AKT signaling pathway. CONCLUSIONS: The synergized promoting role of TEX9 and eIF3b in the progression of ESCC may provide a novel mechanism for exploring viable therapeutic strategies for ESCC.


Asunto(s)
Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Factor 3 de Iniciación Eucariótica/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Factor 3 de Iniciación Eucariótica/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones Desnudos , Proteínas de Neoplasias/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
17.
BMC Cancer ; 19(1): 886, 2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31488089

RESUMEN

BACKGROUND: Predicting lung adenocarcinoma (LUAD) risk is crucial in determining further treatment strategies. Molecular biomarkers may improve risk stratification for LUAD. METHODS: We analyzed the gene expression profiles of LUAD patients from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). We initially used three distinct algorithms (sigFeature, random forest, and univariate Cox regression) to evaluate each gene's prognostic relevance. Survival related genes were then fitted into the least absolute shrinkage and selection operator (LASSO) model to build a risk prediction model for LUAD. After 100,000 times of calculation and model construction, a 16-gene-based prediction model capable of classifying LUAD patients into high-risk and low-risk groups was successfully built. RESULTS: Using a combined strategy, we initially identified 2472 significant survival-related genes. Functional enrichment analysis demonstrated these genes' relevance to tumor initiation and progression. Using the LASSO method, we successfully built a reliable risk prediction model. The risk model was validated in two external sets and an independent set. The expression of these 16 genes was highly correlated with patients' risk. High-risk group patients witnessed poorer recurrence-free survival (RFS) and overall survival (OS) compared to low-risk group patients. Moreover, stratification analysis and decision curve analysis (DCA) confirmed the independence and potential translational value of this predictive tool. We also built a nomogram comprising risk model and stage to predict OS for LUAD patients. CONCLUSIONS: Our risk model may serve as a practical and reliable prognosis predictive tool for LUAD and could provide novel insights into the understanding of the molecular mechanism of this disease.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Aprendizaje Automático , Adenocarcinoma del Pulmón/mortalidad , Anciano , Biomarcadores de Tumor/genética , Bases de Datos Genéticas , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , Tasa de Supervivencia , Transcriptoma/genética
18.
Am J Transl Res ; 11(4): 1997-2012, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31105813

RESUMEN

Lung cancer remains the leading cause of cancer death. DNA methylation plays an essential role in carcinogenesis through regulating gene expression and gene alternative splicing. However, the role of methylation in the tumorigenesis of lung squamous cell carcinoma (SCC) and its association with prognosis remains unclear. Here, we used an integrative approach to evaluate the prognostic value of epigenetic processes in lung SCC by examining the data provided by The Cancer Genome Atlas (TCGA). We found that the mean methylation level was significantly decreased in lung SCC. We also identified methylation-driven genes which were associated with cancer-related pathways. The multivariate Cox regression analysis showed four methylation-driven genes, GCSAM, GPR75, NHLRC1, and TRIM58, could be served as prognostic indicators for lung SCC. Validation on two external GEO datasets showed consistent methylation alterations of the four genes. These findings may have important implications in the understanding of the potential therapeutic method for lung SCC.

19.
Transl Oncol ; 12(4): 646-655, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30818166

RESUMEN

BACKGROUND: Large cell neuroendocrine carcinoma (LCNEC) was categorized into pulmonary neuroendocrine tumors (NETs) according to the World Health Organization classification guideline. However, LCNEC patients often received the chemotherapy regimens similar to non-small cell lung carcinoma (NSCLC) in advanced stage and the therapeutic effect was unsatisfactory. Therefore, this study aimed to investigate the hidden clinical features, prognosis and immunoprofile of the LCNEC, compared with carcinoid and SCLC, to explore whether LCNEC shares similarity with SCLC and potential treatment approaches could be revealed. METHODS: One hundred seventeen pulmonary NETs cases were retrospectively retrieved in this study. The Kaplan-Meier estimator was employed to draw survival curves. Immunohistochemistry was applied to detect NET-related markers expression. RESULTS: In clinical features, compared with carcinoid, LCNEC patients were older, more commonly in male and advanced stage. The parallel phenomena were also found in the high-grade subgroup when compared with the low- to intermediate-grade one. In survival analysis, the 5-year overall survival of LCNECs was 59.1%, which was poorer than that of carcinoids, but better than that of SCLCs. Immunohistochemistry showed that p53 and PTEN functional inactivation, up-regulation of CD117 expression, down-regulation of SSR2A and SSR5 expression were commonly involved in LCNECs when compared with carcinoids, or in the high-grade subgroup when compared with the low- to intermediate-grade one. However, no significant difference was found in the comparison between LCNECs and SCLCs, or NSCLCs and SCLCs. CONCLUSION: In clinical features, survival and immunoprofile, LCNEC showed more similarity with SCLC rather than carcinoid, which might guide novel therapy for pulmonary NETs.

20.
Int J Biol Sci ; 15(1): 127-137, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30662353

RESUMEN

Lung cancer is one of the most frequent malignant tumors, with the top morbidity and mortality, in China. Calpain family regulates cellular processes including migration and invasion. However, the role of Calpain-2 in non-small cell lung cancer (NSCLC) remains unclear. This study aims to explore the bio-function of Calpain-2 on NSCLC and chemoresistance to paclitaxel. In this study, Immunohistochemistry, RT-qPCR and Western blot were performed to detect the Calpain-2 expression and related pathway protein in NSCLC. The Kaplan-Meier product limit estimator and Cox regression were conducted for survival analysis. CCK-8, Transwell, colony-formation, apoptosis and tumor xenograft assays were performed to analyze tumor-promoting role of Calpain-2, and the chemoresistance to paclitaxel. Our data showed that Calpain-2 was up-regulated in NSCLC. Notably, Calpain-2 level positively correlated with differentiation grade and negatively correlated with the 5-year overall survival, which served as an independent prognostic predictor. Knockdown of Calpain-2 inhibited cell proliferation and migration, while promoted apoptosis in vitro. In vivo, Calpain-2-knockdowned cells formed smaller subcutaneous tumors. Meanwhile, knockdown of Calpain-2 down-regulated EGFR and pAKT expression, which weakened the chemoresistance of NSCLC cells to paclitaxel by suppressing cell proliferation and inducing apoptosis, and even enhanced the paclitaxel-mediated downregulation of EGFR and pAKT level. To conclude, Calpain-2 might activate EGFR/pAKT pathway to promote NSCLC progression and contributes to the chemoresistance to paclitaxel, which might be a therapeutic target to prevent or postpone the progression of NSCLC.


Asunto(s)
Calpaína/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Pulmonares/metabolismo , Paclitaxel/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Calpaína/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/genética , Masculino , Ratones Desnudos , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...