Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Mol Med ; 28(12): e18373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38894657

ABSTRACT

Gastric cancer (GC) remains a prominent malignancy that poses a significant threat to human well-being worldwide. Despite advancements in chemotherapy and immunotherapy, which have effectively augmented patient survival rates, the mortality rate associated with GC remains distressingly high. This can be attributed to the elevated proliferation and invasive nature exhibited by GC. Our current understanding of the drivers behind GC cell proliferation remains limited. Hence, in order to reveal the molecular biological mechanism behind the swift advancement of GC, we employed single-cell RNA-sequencing (scRNA-seq) to characterize the tumour microenvironment in this study. The scRNA-seq data of 27 patients were acquired from the Gene Expression Omnibus database. Differential gene analysis, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and Gene Set Enrichment Analysis were employed to investigate 38 samples. The copy number variation level exhibited by GC cells was determined using InferCNV. The CytoTRACE, Monocle and Slingshot analysis were used to discern the cellular stemness and developmental trajectory of GC cells. The CellChat package was utilized for the analysis of intercellular communication crosstalk. Moreover, the findings of the data analysis were validated through cellular functional tests conducted on the AGS cell line and SGC-7901 cell line. Finally, this study constructed a risk scoring model to evaluate the differences of different risk scores in clinical characteristics, immune infiltration, immune checkpoints, functional enrichment, tumour mutation burden and drug sensitivity. Within the microenvironment of GC, we identified the presence of 8 cell subsets, encompassing NK_T cells, B_Plasma cells, epithelial cells, myeloid cells, endothelial cells, mast cells, fibroblasts, pericytes. By delving deeper into the characterization of GC cells, we identified 6 specific tumour cell subtypes: C0 PSCA+ tumour cells, C1 CLDN7+ tumour cells, C2 UBE2C+ tumour cells, C3 MUC6+ tumour cells, C4 CHGA+ tumour cells and C5 MUC2+ tumour cells. Notably, the C2 UBE2C+ tumour cells demonstrated a close association with cell mitosis and the cell cycle, exhibiting robust proliferative capabilities. Our findings were fortified through enrichment analysis, pseudotime analysis and cell communication analysis. Meanwhile, knockdown of the transcription factor CREB3, which is highly active in UBE2C+ tumour cells, significantly impedes the proliferation, migration and invasion of GC cells. And the prognostic score model constructed with CREB3-related genes showcased commendable clinical predictive capacity, thus providing valuable guidance for patients' prognosis and clinical treatment decisions. We have identified a highly proliferative cellular subgroup C2 UBE2C+ tumour cells in GC for the first time. The employment of a risk score model, which is based on genes associated with UBE2C expression, exhibits remarkable proficiency in predicting the prognosis of GC patients. In our investigation, we observed that the knockdown of the transcription factor CREB3 led to a marked reduction in cellular proliferation, migration and invasion in GC cell line models. Implementing a stratified treatment approach guided by this model represents a judicious and promising methodology.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Single-Cell Analysis , Stomach Neoplasms , Tumor Microenvironment , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Tumor Microenvironment/genetics , Cell Proliferation/genetics , Single-Cell Analysis/methods , Cell Line, Tumor , Gene Expression Profiling , DNA Copy Number Variations/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Communication/genetics
2.
Environ Toxicol ; 39(5): 2642-2654, 2024 May.
Article in English | MEDLINE | ID: mdl-38214030

ABSTRACT

BACKGROUND: The exact cause of intracranial aneurysms (IA) is still unclear. However, pro-inflammatory factors are known to contribute to IA progression. The specific changes in the immune microenvironment of IAs remain largely unexplored. METHODS: This study analyzed single-cell sequencing data from a male mouse model of brain aneurysm, focusing on samples before and after elastase-induced Willis aneurysms. The data helped identify eight distinct cell subpopulations: fibroblasts, macrophages, NK cells, endothelial cells, B cells, granulocytes, and monocytes. The study also involved bulk RNA sequencing of 97 IA samples, utilizing ssGSEA and CIBERSORT algorithms for analysis. Intercellular communication among these cells was inferred to understand the immune dynamics in IA. RESULTS: The study found that fibroblasts and macrophages are predominant in various disease states of IA. Notably, the onset of IA was marked by a significant increase in fibroblasts and a decrease in macrophages. There was a marked increase in cellular interactions, especially involving macrophages, at the onset of the disease. Through enrichment analysis, 12 potential immunogenic biomarkers were identified. Of these, Rgs1 emerged as a critical molecule in IA formation, confirmed through secondary validation in a single-cell sequencing dataset. CONCLUSION: This comprehensive analysis of immune cell composition and intercellular communication in IA tissues highlights the significant roles of macrophages and the molecule Rgs1. These findings shed light on the physiological and pathological conditions of IA, offering new insights into its immune microenvironment.


Subject(s)
Intracranial Aneurysm , Mice , Animals , Male , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Endothelial Cells/pathology , Multiomics , Disease Models, Animal , Biomarkers
3.
J Thorac Dis ; 15(2): 552-567, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36910092

ABSTRACT

Background: Surgery combined with chemotherapy (CT) is the best treatment for tumor patients at stage I to IIIA. But there are only few studies specifically evaluated the survival benefits of removing different number of regional lymph nodes (RLNs) for patients with stage IIA non-small cell lung cancer (NSCLC). The objective of this study is to discuss the effect of removing different number of RLNs on survival outcomes in operable patients at stage IIA NSCLC. Methods: Through the use of the Surveillance, Epidemiology, and End Results (SEER) registry, satisfactory patients at stage IIA NSCLC, who had complete clinical information from 2004 to 2015, were identified. Lung cancer-specific survival (LCSS) and overall survival (OS) were compared by the Kaplan-Meier analysis and Cox regression analyses to determine the impact of the confounding factors on the survival outcomes. LCSS and OS as the primary endpoints were compared among patients with different number of RLNs removed. Results: A total of 3,362 patients at stage IIA NSCLC met our criteria, including 173 (5.1%), 486 (14.5%), 2,703 (80.4%) patients without RLNs removed, with 1 to 3 RLNs removed and with greater than or equal to 4 RLNs removed, respectively. Kaplan-Meier survival analyses and Univariate Cox regression analyses revealed that there was a statistically significant difference on survival curve (log rank P<0.001) among the stage IIA NSCLC patients with different number of RLNs removed. Furthermore, multivariable Cox regression analyses on LCSS showed that the hazard ratio (HR) and 95% confidence interval (95% CI) of the 1 to 3 RLNs removed group and greater than or equal to 4 RLNs removed group were 0.622 (0.484-0.800, P<0.001) and 0.545 (0.437-0.680, P<0.001), respectively, compared to without any RLNs removed group. Conclusions: This study illustrated that removing different number of RLNs can affect survival outcomes of operable patients at stage IIA NSCLC. Whether more radical lymphadenectomy is beneficial to patients at stage IIA NSCLC still needs to be researched.

4.
Oncol Res ; 28(9): 945-959, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-34588094

ABSTRACT

Long noncoding RNA LAMTOR5 antisense RNA 1 (LAMTOR5-AS1) has been certified as a risk predictor and diagnostic biomarker of prostate cancer. However, the expression and exact roles of LAMTOR5-AS1 in non-small cell lung cancer (NSCLC) remain unclear. Thus, we measured LAMTOR5-AS1 expression in NSCLC and gauged its clinical value. The detailed roles and downstream working mechanism of LAMTOR5-AS1 in NSCLC were comprehensively unraveled. qRT-PCR was applied to measure gene expression. Functionally, utilizing small interfering RNA, LAMTOR5-AS1 was ablated, and the functional alterations were addressed by means of different experiments. The targeting activities between LAMTOR5-AS1 and microRNA-506-3p (miR-506-3p) and between miR-506-3p and E2F transcription factor 6 (E2F6) were confirmed by RNA immunoprecipitation and luciferase reporter assays. LAMTOR5-AS1 overexpression in NSCLC was verified in TCGA datasets and our own cohort and manifested an evident relationship with poor prognosis. Interference with LAMTOR5-AS1 led to repression of the proliferation, cloning, and metastasis abilities of NSCLC cells in vitro. We further confirmed an obvious increase in LAMTOR5-AS1-silenced NSCLC cell apoptosis. Furthermore, the absence of LAMTOR5-AS1 restricted tumor growth in vivo. Mechanistically, LAMTOR5-AS1 sponged miR-506-3p in NSCLC cells. Furthermore, E2F6, a downstream target of miR-506-3p, was under the control of LAMTOR5-AS1, which was realized by decoying miR-506-3p. Rescue experiments showed that miR-506-3p suppression or E2F6 reintroduction was capable of remitting LAMTOR5-AS1 deficiency-triggered anticarcinogenic actions in NSCLC. Our study confirmed the exact roles of LAMTOR5-AS1 for the first time and revealed that LAMTOR5-AS1 knockdown disrupts the malignancy of NSCLC by targeting the miR-506-3p/E2F6 axis. Targeting the LAMTOR5-AS1/miR-506-3p/E2F6 pathway may be instrumental for managing patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation , E2F6 Transcription Factor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Male , MicroRNAs/genetics , RNA, Long Noncoding/genetics
5.
Chin Med J (Engl) ; 134(18): 2166-2174, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34553699

ABSTRACT

BACKGROUND: The effectiveness of bronchial thermoplasty (BT) has been reported in patients with severe asthma. This study compared the effects of BT and cryoballoon ablation (CBA) therapy on the airway smooth muscle (ASM). METHODS: Eight healthy male beagle dogs were included in this experiment. In the preliminary experiment, one dog received BT treatment for both lower lobe bronchus, another dog received CBA treatment for 7 s on the upper and lower lobe of right bronchus, and 30 s on the left upper and lower lobe. The treatments were performed twice at an interval of 1 month. In subsequent experiments, the right lower lobe bronchus was treated with BT, and the left lower lobe bronchus was treated with CBA. The effects of treatment were observed after 1 (n = 3) month and 6 months (n = 3). Hematoxylin-eosin staining, Masson trichrome staining, and immunohistochemical staining were used to compare the effects of BT and CBA therapy on the ASM thickness, collagen fibers synthesis, and M3 receptor expression after treatment. One-way analysis of variance with Dunnett post hoc test was used to analyze the differences among groups. RESULTS: In the preliminary experiment, the ASM ablation effect of 30-s CBA was equivalent to that of 7-s CBA (ASM thickness: 30.52 ±â€Š7.75 µm vs. 17.57 ±â€Š15.20 µm, P = 0.128), but the bronchial mucociliary epithelium did not recover, and large numbers of inflammatory cells had infiltrated the mucosal epithelium at 1-month post-CBA with 30-s freezing. Therefore, we chose 7 s as the CBA treatment time in our follow-up experiments. Compared with the control group (35.81 ±â€Š11.02 µm), BT group and CBA group (13.41 ±â€Š4.40 µm and 4.81 ±â€Š4.44 µm, respectively) had significantly decreased ASM thickness after 1 month (P < 0.001). Furthermore, the ASM thickness was significantly lower in the 1-month post-CBA group than in the 1-month post-BT group (P = 0.015). There was no significant difference in ASM thickness between the BT and CBA groups after six months (9.92 ±â€Š4.42 µm vs. 7.41 ±â€Š7.20 µm, P = 0.540). Compared with the control group (0.161 ±â€Š0.013), the average optical density of the ASM M3 receptor was significantly decreased in 6-month post-BT, 1-month post-CBA, and 6-month post-CBA groups (0.070 ±â€Š0.022, 0.072 ±â€Š0.012, 0.074 ±â€Š0.008, respectively; all P < 0.001). There was no significant difference in the average optical density of ASM M3 receptor between the BT and CBA therapy groups after six months (P = 0.613). CONCLUSIONS: CBA therapy effectively ablates the ASM, and its ablation effect is equivalent to that of BT with a shorter onset time. A neural mechanism is involved in both BT and CBA therapy.


Subject(s)
Bronchial Thermoplasty , Cryosurgery , Animals , Bronchi/surgery , Bronchoscopy , Dogs , Humans , Male , Muscle, Smooth
6.
J Cancer ; 12(6): 1708-1714, 2021.
Article in English | MEDLINE | ID: mdl-33613758

ABSTRACT

Objectives: We performed this study to compare survival outcomes of segmentectomy (SG) and wedge resection (WR) in stage IA lung squamous cell carcinoma (SQCC) and lung adenocarcinoma (AD). Methods: Using the Surveillance, Epidemiology, and End Results registry (SEER), we identified 1529 and 4070 patients with stage IA SQCC and AD, respectively, who had complete clinical information between 2004 and 2015. We used Kaplan-Meier analysis to determine the propensity score for patients with limited resection based on the preoperative characteristics of patients. Lung cancer-specific survival (LCSS) was compared in patients treated with WR and SG after adjusting, stratifying, or matching lung cancer patients according to propensity score. Results: Kaplan-Meier analysis demonstrated that there was a statistically significant difference in survival curves (log rank P=0.01) for patients with stage IA SQCC between SG and WR. But there was no statistically significant difference in survival curves (log rank P>0.05) in patients with stage IA AD between the two limited resections. Compared with the WR, The hazard ratios (95% confidence intervals) of SG were 0.689 (0.519-0.914) and 0.896 (0.752-1.067) in patients with stage IA SQCC and AD, respectively. Conclusion: This study suggests that SG can yield superior survival outcome compared with WR in patients with stage IA SQCC. However, the survival outcomes of SG and WR are generally equivalent in patients with stage IA AD.

7.
J Cancer ; 11(6): 1634-1640, 2020.
Article in English | MEDLINE | ID: mdl-32047569

ABSTRACT

Purpose: To compare the survival outcomes of ablation and stereotactic body radiotherapy (SBRT) in inoperable patients with stage IA non-small cell lung cancer (NSCLC). Patients and Methods: Using the Surveillance, Epidemiology, and End Results (SEER) database, we identified 6,395 patients with stage IA NSCLC who had complete clinical information from 2004 to 2015. Kaplan-Meier analysis was performed to determine the propensity score based on the clinical characteristics of patients with stage IA NSCLC. Overall survival (OS) was compared between patients with stage IA NSCLC who were treated with ablation and SBRT after adjusting, stratifying, or matching. Results: Kaplan-Meier analysis demonstrated no significant difference in survival curves (log-rank, p>0.05) between the ablation and SBRT groups. Compared with the SBRT group, the hazard ratio (HR) (95% confidence interval [CI]) of OS was 0.930 (0.817-1.058, p=0.269) in the ablation group on univariate analysis. On multivariate analysis, similar effects on OS (HR: 0.974, 95% CI: 0.858-1.105, p=0.680) were seen in patients with stage IA NSCLC in both the groups. Conclusions: This study suggests that survival does not differ significantly between patients with stage IA NSCLC treated with ablation and SBRT. These results will be helpful for patients with stage IA NSCLC who are ineligible for surgery.

8.
Cancer Manag Res ; 11: 7089-7101, 2019.
Article in English | MEDLINE | ID: mdl-31440096

ABSTRACT

PURPOSE: Currently, systemic chemotherapy combined with thoracic radiation is the standard treatment for patients with small-cell lung cancer (SCLC). However, the treatment of early stage SCLC remains controversial. This study evaluated the survival outcomes of surgical treatments and the effect of adjuvant chemotherapy and radiotherapy on lung cancer-specific survival (LCSS) in patients with early stage SCLC. METHODS: Using the Surveillance, Epidemiology, and End Results registry, we identified 2,453 patients with early stage SCLC (1,295 women and 1,158 men) who had complete clinical information between 2004 and 2015. The Kaplan-Meier analysis was used to determine the propensity score based on the characteristics of patients with early stage SCLC. LCSS was compared between patients treated with surgery and non-surgery after adjusting, stratifying, or matching patients with early stage SCLC. In addition, we compared the effects of chemotherapy and radiotherapy on LCSS in patients with early stage SCLC. RESULTS: Overall, 687 (28.0%) and 1,766 (72.0%) patients with early stage SCLC did and did not undergo surgery, respectively. Kaplan-Meier analysis demonstrated a statistically significant difference in survival curves between the surgery and non-surgery groups (log-rank p<0.001). Compared with the non-surgery group, the LCSS of the surgery group was better (hazard ratio [HR]:0.494, 95% confidence interval [CI]:0.415-0.587, p<0.001) in patients with early stage SCLC when using a Cox model for multivariate analysis. There was no statistically significant difference (p=0.847) in LCSS between patients with early stage SCLC with and without chemotherapy in the multivariate analysis. Radiotherapy had favorable effects on LCSS (HR: 0.579, 95% CI: 0.500-0.671, p<0.001) in patients with early stage SCLC using multivariate analysis. CONCLUSIONS: Our study results suggest that LCSS conferred by surgery was higher than that conferred by non-surgery and that radiotherapy is associated with better survival in patients with early stage SCLC. This study findings should be confirmed in prospective studies.

SELECTION OF CITATIONS
SEARCH DETAIL
...