Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Cancer Lett ; 581: 216498, 2024 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-38029539

RESUMO

Abnormal regulation of RNA binding proteins (RBPs) plays an essential role in tumorigenesis and progression, but their functions and mechanisms remain largely elusive. Previously, we reported that Pumilio 1 (PUM1), a RBP, could regulate glycolysis metabolism and promote the progression of gastric cancer (GC). However, the role of PUM1 in tumor immune regulation remains largely elusive. In this study, we report that PUM1 induces immune escape through posttranscriptional regulation of PD-L1 in GC. We used multiplexed immunohistochemistry to analyze the correlation between PUM1 expression and immune microenvironment in GC. The effect of PUM1 deficiency on tumor killing of T cells was examined in vitro and in vivo. The molecular mechanism of PUM1 was evaluated via RNA immunoprecipitation, chromatin immunoprecipitation, Western blot, co-immunoprecipitation, and RNA stability assays. Clinically, elevated PUM1 expression is associated with high-expression of PD-L1, lack of CD8+ T cell infiltration and poor prognosis in GC patients. PUM1 positively regulates PD-L1 expression and PUM1 reduction enhances T cell killing of tumors. Mechanistically, PUM1 directly binds to nucleophosmin/nucleoplasmin 3 (NPM3) mRNA and stabilizes NPM3. NPM3 interacts with NPM1 to promote NPM1 translocation into the nucleus and increase the transcription of PD-L1. PUM1 inhibits the anti-tumor activity of T cells through the PUM1/NPM3/PD-L1 axis. In summary, this study reveals the critical post-transcriptional effect of PUM1 in the modulation of PD-L1-dependent GC immune escape, thus provides a novel indicator and potential therapeutic target for cancer immunotherapy.


Assuntos
Neoplasias Gástricas , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleoplasminas/metabolismo , Proteínas de Ligação a RNA/genética , Neoplasias Gástricas/patologia , Microambiente Tumoral
3.
Front Immunol ; 14: 1140328, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37180146

RESUMO

Introduction: Gastric cancer (GC) is the fifth most common tumor, contributing to the third-highest number of cancer-related deaths. Hypoxia is a major feature of the tumor microenvironment. This study aimed to explore the influence of hypoxia in GC and establish a hypoxia-related prognostic panel. Methods: The GC scRNA-seq data and bulk RNA-seq data were downloaded from the GEO and TCGA databases, respectively. AddModuleScore() and AUCell() were used to calculate module scores and fractions of enrichment for hypoxia-related gene expression in single cells. Least absolute shrinkage and selection operator cox (LASSO-COX) regression analysis was utilized to build a prognostic panel, and hub RNAs were validated by qPCR. The CIBERSORT algorithm was adopted to evaluate immune infiltration. The finding of immune infiltration was validated by a dual immunohistochemistry staining. The TIDE score, TIS score and ESTIMATE were used to evaluate the immunotherapy predictive efficacy. Results: Hypoxia-related scores were the highest in fibroblasts, and 166 differentially expressed genes were identified. Five hypoxia-related genes were incorporated into the hypoxia-related prognostic panel. 4 hypoxia-related genes (including POSTN, BMP4, MXRA5 and LBH) were significantly upregulated in clinical GC samples compared with the normal group, while APOD expression decreased in GC samples. Similar results were found between cancer-associated fibroblasts (CAFs) and normal fibroblasts (NFs). A high hypoxia score was associated with advanced grade, TNM stage, N stage, and poorer prognosis. Decreased antitumor immune cells and increased cancer-promoting immune cells were found in patients with high hypoxia scores. Dual immunohistochemistry staining showed high expression of CD8 and ACTA2 in gastric cancer tissue. In addition, the high hypoxia score group possessed higher TIDE scores, indicating poor immunotherapy benefit. A high hypoxia score was also firmly related to sensitivity to chemotherapeutic drugs. Discussion: This hypoxia-related prognostic panel may be effective in predicting the clinical prognosis, immune infiltrations, immunotherapy, and chemotherapy in GC.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Prognóstico , Hipóxia/genética , Algoritmos , Análise de Sequência de RNA , Microambiente Tumoral/genética
4.
Exp Cell Res ; 423(1): 113453, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36584745

RESUMO

BACKGROUND: CYRI-B plays key roles in regulating cell motility in nontumor cells. However, the role and function of CYRI-B have rarely been studied in cancer cells, including gastric cancer. The purpose of this study was to investigate the clinical significance, biological function and underlying molecular mechanism of CYRI-B in gastric cancer. METHOD: CYRI-B protein levels were detected by immunohistochemistry (IHC) and western blotting (WB). Gastric cancer cells and organoid models were evaluated to explore the correlation of CYRI-B with collagen type I. The function of CYRI-B in proliferation, migration, invasion in gastric cancer was evaluated by in vitro and in vivo experiments. RESULT: CYRI-B protein levels were downregulated in gastric cancer. Low expression of CYRI-B was related to later tumor stage and poorer prognosis. CYRI-B expression was reduced when cells were cultured in collagen type I, which was mediated by collagen receptor DDR1. Knockdown of CYRI-B promoted migration, invasion and EMT in vivo and in vitro. Mechanistically, knockdown of CYRI-B activated the Rac1-STAT3 pathway. CONCLUSION: Our findings showed that CYRI-B plays an important role in the tumor microenvironment, and is associated with malignant characteristics acquired by gastric cancer. This study may provide new targets for future therapeutic interventions for tumor metastasis.


Assuntos
Neoplasias Gástricas , Humanos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Colágeno Tipo I/metabolismo , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , Invasividade Neoplásica/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/patologia , Microambiente Tumoral , Proteínas Mitocondriais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
5.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36572651

RESUMO

Immune checkpoint inhibitors (ICI) show high efficiency in a small fraction of advanced gastric cancer (GC). However, personalized immune subtypes have not been developed for the prediction of ICI efficiency in GC. Herein, we identified Pan-Immune Activation Module (PIAM), a curated gene expression profile (GEP) representing the co-infiltration of multiple immune cell types in tumor microenvironment of GC, which was associated with high expression of immunosuppressive molecules such as PD-1 and CTLA-4. We also identified Pan-Immune Dysfunction Genes (PIDG), a conservative PIAM-derivated GEP indicating the dysfunction of immune cell cooperation, which was associated with upregulation of metastatic programs (extracellular matrix receptor interaction, TGF-ß signaling, epithelial-mesenchymal transition and calcium signaling) but downregulation of proliferative signalings (MYC targets, E2F targets, mTORC1 signaling, and DNA replication and repair). Moreover, we developed 'GSClassifier', an ensemble toolkit based on top scoring pairs and extreme gradient boosting, for population-based modeling and personalized identification of GEP subtypes. With PIAM and PIDG, we developed four Pan-immune Activation and Dysfunction (PAD) subtypes and a GSClassifier model 'PAD for individual' with high accuracy in predicting response to pembrolizumab (anti-PD-1) in advance GC (AUC = 0.833). Intriguingly, PAD-II (PIAMhighPIDGlow) displayed the highest objective response rate (60.0%) compared with other subtypes (PAD-I, PIAMhighPIDGhigh, 0%; PAD-III, PIAMlowPIDGhigh, 0%; PAD-IV, PIAMlowPIDGlow, 17.6%; P = 0.003), which was further validated in the metastatic urothelial cancer cohort treated with atezolizumab (anti-PD-L1) (P = 0.018). In all, we provided 'GSClassifier' as a refined computational framework for GEP-based stratification and PAD subtypes as a promising strategy for exploring ICI responders in GC. Metastatic pathways could be potential targets for GC patients with high immune infiltration but resistance to ICI therapy.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Aprendizado de Máquina , Microambiente Tumoral
6.
Int J Biol Sci ; 18(16): 6068-6083, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439876

RESUMO

O-glycosylation is a widespread post-translational modification of proteins. Aberrant O-glycosylation is a hallmark of cancer. Here, we show that the polypeptide N-acetylgalactosamine-transferase 1 (GALNT1) is frequently upregulated in gastric cancer and is correlated with poor survival. Overexpression of GALNT1 promoted, whereas knockdown suppressed proliferation, migration, and invasion of gastric cancer cells in vitro and in vivo. Mechanistically, GALNT1 enhances aberrant initiation of O-glycosylation and results in CD44 glycoproteins modified with abundant Tn antigens, thereby activating the Wnt/ß-catenin signaling pathway. Collectively, this study demonstrates that GALNT1 overexpression in gastric cancer promotes the Wnt/ß-catenin signaling pathway via abnormal O-glycosylation of CD44 to enhance malignancy, providing a novel strategy for the development of therapeutic reagents against gastric cancer.


Assuntos
Neoplasias Gástricas , Via de Sinalização Wnt , Humanos , Via de Sinalização Wnt/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Glicosilação , Fenótipo , Receptores de Hialuronatos/genética
7.
Front Oncol ; 12: 918719, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36081567

RESUMO

Fibronectin 1 (FN1) is a glycoprotein found throughout the extracellular matrix that has a role in the onset and progression of cancer. However, its immune relationship with gastric cancer is still unclear. FN1 was systematically reviewed by Gene Expression Profiling Interactive Analysis (GEPIA), Linked Omics, Tumor IMmune Estimation Resource (TIMER), and Kaplan-Meier (KM) plotter analysis. The TIMER, GEPIA, TISIDB, and cBioPortal databases investigated the association of FN1 with tumor immune infiltration and validated using immunohistochemistry. We discovered that tumor tissue expresses FN1 at a higher level than neighboring tissue, and those genes coexpressed with FN1 have a poor prognosis. At the same time, we discovered that increased FN1 expression was related to immunological infiltration, particularly macrophage infiltration. Using immunohistochemistry, we discovered that FN1 expression was tightly connected to M2 macrophages. It can be concluded that FN1 can affect the immunological microenvironment and is a prognostic marker in gastric cancer.

8.
Front Immunol ; 13: 922138, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36090985

RESUMO

The Schlafen (SLFN) gene family plays an important role in immune cell differentiation and immune regulation. Previous studies have found that the increased SLFN5 expression in patients with intestinal metaplasia correlates with gastric cancer (GC) progression. However, no investigation has been conducted on the SLFN family in GC. Therefore, we systematically explore the expression and prognostic value of SLFN family members in patients with GC, elucidating their possible biological function and its correlation with tumor immune cells infiltration. TCGA database results indicated that the SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN13 expression was significantly higher in GC. The UALCAN and KM plotter databases indicated that enhanced the SLFN family expression was associated with lymph node metastasis, tumor stage, and tumor grade and predicted an adverse prognosis. cBioportal database revealed that the SLFN family had a high frequency of genetic alterations in GC (about 12%), including mutations and amplification. The GeneMANIA and STRING databases identified 20 interacting genes and 16 interacting proteins that act as potential targets of the SLFN family. SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN14 may be implicated in the immunological response, according to Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Additionally, Timer and TISIDB databases indicate that SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN14 are involved in the immune response. Furthermore, Timer, TCGA, and TISIDB databases suggested that the SLFN5, SLFN11, SLFN12, SLFN12L, and SLFN14 expression in GC is highly linked with immune cell infiltration levels, immune checkpoint, and the many immune cell marker sets expression. We isolated three samples of peripheral blood mononuclear cell (PBMC) and activated T cells; the results showed the expression of SLFN family members decreased significantly when T cell active. In conclusion, the SLFN family of proteins may act as a prognostic indicator of GC and is associated with immune cell infiltration and immune checkpoint expression in GC. Additionally, it may be involved in tumor immune evasion by regulating T cell activation.


Assuntos
Neoplasias Gástricas , Proteínas de Ciclo Celular/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Metaplasia , Proteínas Nucleares , Prognóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
9.
Front Med (Lausanne) ; 9: 829033, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721089

RESUMO

Background: Mucinous appendiceal adenocarcinoma (MAA) is a rare, heterogeneous disease. Patients with unrespectable mucinous appendiceal adenocarcinoma presenting with peritoneal spread are treated by intraperitoneal chemotherapy, hyperthermic intraperitoneal chemotherapy, systemic chemotherapy, or targeted therapy. However, there are no guidelines for efficacious drugs against mucinous appendiceal adenocarcinoma. Therefore, relevant high-fidelity models should be investigated to identify effective drugs for individual therapy. Methods: Surgical tumor specimens were obtained from a mucinous appendiceal adenocarcinoma patient. The tissue was digested and organoid culture was established. H&E and immunohistochemistry staining as well as DNA sequencing was performed on tissue and organoid. The pathological characteristics and gene mutations of the organoid were compared to those of the original tumor. Drug sensitivity tests were performed on organoid and the patient clinical responds to chemotherapy and targeted therapy was compared. Results: Organoids were successfully established and stably passaged. Pathological characteristics of organoids including H&E staining and expression of protein markers (CK20, CDX-2, STAB2, CD7, PAX8) were consistent to those of the original tumor. Moreover, the organoids carried the same gene mutations as the primary tumor. Sensitivity of the organoids to chemotherapeutic drugs and tyrosine kinase inhibitors included: 5-FU (IC50 43.95 µM), Oxaliplatin (IC50 23.49 µM), SN38 (IC50 1.02 µM), Apatinib (IC50 0.10 µM), Dasatinib (IC50 2.27 µM), Docetaxel (IC50 5.26 µM), Regorafenib (IC50 18.90 µM), and Everolimus (IC50 9.20 µM). The sensitivities of organoid to these drugs were comparable to those of the patient's clinical responses. Conclusion: The mucinous appendiceal adenocarcinoma organoid model which retained the characteristics of the primary tumor was successfully established. Combined organoid-based drug screening and high throughput sequencing provided a promising way for mucinous appendiceal adenocarcinoma treatment.

10.
J Cancer ; 13(7): 2126-2137, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35517410

RESUMO

Most colorectal cancer (CRC) patients are insensitive to immune checkpoint inhibitors (ICIs) due to the immunosuppressive tumor microenvironment (TME). Epigenetic factors such as the bromo-and extraterminal domain (BET) family proteins may be responsible for the immunosuppressive microenvironment. Previous studies have shown that inhibitors of BET family proteins have the potential to remodel the immunosuppressive TME. However, data on the role of BET inhibitors in immune microenvironment in CRC remains unclear. Here, we evaluated the immunoregulatory role of JQ1, a BET inhibitor, in CRC. Transcriptome sequencing data showed that JQ1 decreased CD274 expression and increased H2Kb expression in MC38 cells. Flow cytometry assays demonstrated that JQ1 decreased cell-surface PD-L1 expression in MC38 and HCT116 cells. Moreover, JQ1 significantly increased cell-surface expression of major histocompatibility complex class I (MHC-I) in MC38 cells and HCT116 cells. Antigen-specific cytotoxic T lymphocytes (CTLs) assay demonstrated that JQ1 enhanced the MHC-I-mediated cytotoxicity of CTLs. Mouse colon cancer cell line MC38 was used to establish the syngeneic mouse tumor model. Compared with the control, JQ1 significantly inhibited tumor growth and prolonged the overall survival of the mice. Besides, JQ1 did not only inhibit tumor growth by enhancing anti-tumor immunity, but also promoted the anti-tumor effect of PD-1 antibody. In addition, our data showed that JQ1 reduced infiltration of intratumoral regulatory T cells (Treg), thus remodeling the immunosuppressive TME. Taken together, these results highlight a new approach that enhances anti-PD-1 sensitivity in CRC.

11.
Front Cell Dev Biol ; 10: 808212, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35399517

RESUMO

Integrin alpha L (ITGAL) is a member of the integrin family in which the abnormal expression is linked with carcinogenesis and immune regulation. However, the relation between ITGAL and the prognosis of gastric cancer (GC) and tumor-infiltrating lymphocytes (TILs) are not well understood. The differential expressions of ITGAL in human tumors and the clinical prognosis in GC were systematically analyzed via multiple databases including Gene Expression Profiling Interaction Analysis (GEPIA), UALCAN, Tumor Immune Estimation Resource (TIMER), and Kaplan-Meier (KM) plotter. TIMER, GEPIA, and TISIDB databases were used to comprehensively investigate the correlation between ITGAL and tumor infiltration immune cells. Also, further results were investigated by immunohistochemistry, qRT-PCR, and Western blot. We found that ITGAL expression in GC samples was considerably increased than in peritumor samples. Sample type, subgroup, cancer stage, lymphatic node stage, and worse survival were strongly related to high ITGAL expression. Moreover, upregulated ITGAL expression was strongly connected with immunomodulators, chemokines, and infiltrating levels of CD8+, CD4+ T cell, B cell, monocyte, neutrophil, macrophage, T-cell regulatory, NK cell, and myeloid dendritic cell in stomach adenocarcinoma (STAD). Specifically, immunohistochemistry and bioinformatic analysis showed that ITGAL expression was shown to have strong relationships with various immunological marker sets including PD1 (T-cell exhaustion marker). In conclusion, ITGAL is a prognostic biomarker for GC patients. It might regulate tumor immune microenvironment leading to poor prognosis. Furthermore, studies are essential to explore therapeutic targeting ITGAL.

12.
Front Pharmacol ; 13: 759247, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35370669

RESUMO

Objective: Metabolic acidosis often occurs in the paediatric intensive care unit (PICU). Although sodium bicarbonate (SB) has been widely used in paediatrics, data on the effect of SB on children with metabolic acidosis in the PICU are scarce. Methods: Patients with metabolic acidosis who were treated with SB within 48 h of PICU admission were screened. Multivariate logistic regression, subgroup analysis, and propensity score matching (PSM) were used to investigate the relationships between SB infusion and clinical outcomes. Results: A total of 1,595 patients with metabolic acidosis were enrolled in this study. In the multivariate logistic regression model, SB infusion was not correlated with in-hospital mortality (odds ratio (OR) 0.87, 95% confidence interval (CI) 0.47-1.63, p = 0.668), but was significantly correlated with hypernatraemia (OR 1.98, 95% CI 1.14-3.46, p = 0.016), hypokalaemia (OR 2.01, 95% CI 1.36-2.96, p < 0.001), and hypocalcaemia (OR 4.29, 95% CI 2.92-6.31, p < 0.001). In the pH value, lactate level, acute kidney injury level, age grouping, and anion gap level subgroups, the ORs for SB and in-hospital mortality were not statistically significant. After PSM, the results remained unchanged. Conclusion: SB infusion does not reduce the in-hospital mortality of severely ill children with metabolic acidosis and increases the risk of hypernatraemia, hypokalaemia, and hypocalcaemia. More effort should be focused on eliminating the causes of metabolic acidosis rather than SB infusion.

14.
Cancers (Basel) ; 15(1)2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36612024

RESUMO

Syntaxin-6 (STX6), a vesicular transport protein, is a direct target of the tumor suppressor gene P53, supporting cancer growth dependent on P53. However, STX6's function in the tumor microenvironment has yet to be reported. In this research, we comprehensively explored the role of the oncogene STX6 in pan-cancer by combining data from several databases, including the Cancer Genome Atlas, CPTAC, cBioPortal, and TIMER. Then, we verified the carcinogenic effect of STX6 in hepatocellular carcinoma (HCC) and colorectal cancer (CRC) through a series of experiments in vitro and in vivo. Bioinformatics analysis demonstrated that STX6 is an oncogene for several cancers and is mainly involved in the cell cycle, epithelial-mesenchymal transition, oxidative phosphorylation, and tumor immune modulation, especially for tumor-associated fibroblasts (CAFs) and NKT cells. Additionally, a high level of STX6 could indicate patients' resistance to immunotherapy. Our own data indicated that the STX6 level was upregulated in HCC and CRC. Knockdown of the STX6 levels could arrest the cell cycle and restrain cell proliferation, migration, and invasion. RNA-seq indicated that STX6 was significantly involved in pathways for cancer, such as the MAPK signal pathway. In a mouse model, knockdown of STX6 inhibited tumor growth and potentiated anti-PD-1 efficacy. In light of the essential roles STX6 plays in carcinogenesis and cancer immunology, it has the potential to be a predictive biomarker and a target for cancer immunotherapy.

15.
Curr Oncol ; 30(1): 171-183, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36661663

RESUMO

BACKGROUND: It is widely acknowledged that the molecular biological characteristics of diffuse-type gastric cancer are different from intestinal-type gastric cancer. Notwithstanding that significant progress in high-throughput sequencing technology has been made, there is a paucity of effective prognostic biomarkers for diffuse gastric cancer for clinical practice. METHODS: We downloaded four GEO datasets (GSE22377, GSE38749, GSE47007 and GSE62254) to establish and validate a prognostic two-gene signature for diffuse gastric cancer. The TGCA-STAD dataset was used for external validation. The optimal gene signature was established by using Cox regression analysis. Receiver operating characteristic (ROC) methodology was used to find the best prognostic model. Gene set enrichment analysis was used to analyze the possible signaling pathways of the two genes (MEF2C and TRIM15). RESULTS: A total of four differently expressed genes (DEGs) (two upregulated and two downregulated) were identified. After a comprehensive analysis, two DEGs (MEF2C and TRIM15) were utilized to construct a prognostic model. A prognostic prediction model was constructed according to T stage, N stage, M stage and the expression of MEF2C and TRIM15. The area under the time-dependent receiver operator characteristic was used to evaluate the performance of the prognosis model in the GSE62254 dataset. CONCLUSIONS: We demonstrated that MEF2C and TRIM15 might be key genes. We also established a prognostic nomogram based on the two-gene signature that yielded a good performance for predicting overall survival in diffuse-type gastric cancer.


Assuntos
Nomogramas , Neoplasias Gástricas , Humanos , Prognóstico , Neoplasias Gástricas/genética , Sequenciamento de Nucleotídeos em Larga Escala
16.
J Exp Clin Cancer Res ; 40(1): 260, 2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34412665

RESUMO

BACKGROUND: Oxaliplatin is one of the most commonly used chemotherapeutic agent for the treatment of various cancers, including gastric cancer. It has, however, a narrow therapeutic index due to its toxicity and the occurrence of drug resistance. Hence, it is of great significance to develop novel therapies to potentiate the anti-tumor effect and reduce the toxicity of oxaliplatin. In our previous study, we demonstrated that ethaselen (BBSKE), an inhibitor of thioredoxin reductase, effectively inhibited the growth of gastric cancer cells and promoted apoptosis in vitro. In the present study, we investigated whether BBSKE can potentiate the anti-tumor effect of oxaliplatin in gastric cancer in vivo and vitro. METHODS: Cellular apoptosis and ROS levels were analyzed by flow cytometry. Thioredoxin reductase 1 (TrxR1) activity in gastric cancer cells, organoid and tumor tissues was determined by using the endpoint insulin reduction assay. Western blot was used to analyze the expressions of the indicated proteins. Nude mice xenograft models were used to test the effects of BBSKE and oxaliplatin combinations on gastric cancer cell growth in vivo. In addition, we also used the combined treatment of BBSKE and oxaliplatin in three cases of gastric cancer Patient-Derived organoid (GC-PDO) to detect the anti-tumor effect. RESULTS: We found that BBSKE significantly enhanced oxaliplatin-induced growth inhibition in gastric cancer cells by inhibiting TrxR1 activity. Because of the inhibition of TrxR1 activity, BBSKE synergized with oxaliplatin to enhance the production of ROS and activate p38 and JNK signaling pathways which eventually induced apoptosis of gastric cancer cells. In vivo, we also found that BBSKE synergized with oxaliplatin to suppress the gastric cancer tumor growth in xenograft nude mice model, accompanied by the reduced TrxR1 activity. Remarkably, we found that BBSKE attenuated body weight loss evoked by oxaliplatin treatment. We also used three cases of GC-PDO and found that the combined treatment of BBSKE and oxaliplatin dramatically inhibited the growth and viability of GC-PDO with increased ROS level, decreased TrxR1 activity and enhanced apoptosis. CONCLUSIONS: This study elucidates the underlying mechanisms of synergistic effect of BBSKE and oxaliplatin, and suggests that the combined treatment has potential value in gastric cancer therapy.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Organosselênicos/farmacologia , Oxaliplatina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/metabolismo , Tiorredoxina Redutase 1/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sinergismo Farmacológico , Ativação Enzimática , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Nus , Organoides , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Cancer Res Clin Oncol ; 146(11): 2937-2947, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32451690

RESUMO

PURPOSE: To investigate the clinicopathological features and survival outcomes between young and old patients with gastric cancer (GC), and further determine the role of young age in the prognosis of GC. METHODS: Patients with stage I-III gastric adenocarcinomas undergoing curative surgery were enrolled, divided into young (aged 18-49 years, YG), middle-aged (50-59 years, MG), and old (≥ 60 years, OG) groups. Exclusion criteria were neoadjuvant therapy and history of malignant tumors. Clinicopathological features, overall survival (OS), disease-free survival (DFS), and recurrence patterns were compared among three groups. RESULTS: 1131 patients were finally included, with 270, 314, and 547 cases in the YG, MG, and OG, respectively. Compared to others, YG had higher proportion of female, middle-third gastric cancer, poor differentiation, N3b stage, and adjuvant chemotherapy. YG demonstrated poorer 5-year OS than MG (62.4% vs. 70.8%, P = 0.019), but better than OG (62.4% vs. 58.7%, P = 0.031). YG also suffered inferior 5-year DFS (75.2% vs. 82.8%, P = 0.040) compared with MG, and higher incidence of peritoneal recurrence than MG (15.1% vs. 5.2%, P < 0.001) and OG (15.1% vs. 4.1%, P < 0.001). Multivariate analysis identified young age as the independent prognostic factor for OS [hazard ratio (HR) = 1.347, 95% CI 1.018-1.781, P = 0.037], DFS (HR = 1.601, 95% CI 1.079-2.376, P = 0.019), and peritoneal recurrence (HR = 2.936, 95% CI 1.505-5.726, P = 0.002). CONCLUSIONS: Young GC patients demonstrated aggressive features with poor prognosis and enhanced management may be warranted for this subgroup.


Assuntos
Adenocarcinoma/patologia , Neoplasias Gástricas/patologia , Adenocarcinoma/mortalidade , Adolescente , Adulto , Fatores Etários , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Gástricas/mortalidade , Adulto Jovem
18.
J Gastrointest Surg ; 23(7): 1474-1484, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30617772

RESUMO

BACKGROUND: Rectal cancers have long been treated as a single-entity disease; however, whether the prognosis of high rectal cancer (inferior margin located 10.1 to 15.0 cm from the anal verge) differs from that of mid/low rectal cancer (0 to 10.0 cm) remains disputed. METHODS: Patients with stages I-III rectal adenocarcinomas undergoing curative-intent surgery were enrolled between 2007 and 2013 in this retrospective analysis. Exclusion criteria were neoadjuvant therapy or concurrent cancers. Propensity score matching and Cox regression analysis were performed to compare a 5-year overall and cancer-specific survival between patients with high and mid/low rectal cancer. RESULTS: Of 613 patients who met the inclusion criteria, 199 (32.5%) and 414 (67.5%) had high and mid/low rectal cancer, respectively. After propensity score matching (187 cases for each group), the high group showed a better overall survival (70.9 vs. 56.9%, p = 0.042) and cancer-specific survival (77.4 vs. 60.3%, p = 0.028) at 5 years compared with the mid/low group with stage III disease. However, high rectal cancer did not demonstrate prognostic superiority in stages I-II disease. Multivariate analysis identified high tumor location as an independent prognostic factor for cancer-specific survival (hazards ratio = 0.422, 95% confidence interval 0.226-0.786, p = 0.007) and overall survival (hazards ratio = 0.613, 95% confidence interval 0.379-0.991, p = 0.046). CONCLUSIONS: Patients with stage III high rectal adenocarcinoma demonstrated better overall and cancer-specific survival than those with mid/low type, and tumor location was an independent prognostic factor for patients with rectal carcinomas.


Assuntos
Adenocarcinoma/cirurgia , Neoplasias Retais/patologia , Neoplasias Retais/cirurgia , Reto/patologia , Adenocarcinoma/secundário , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Pontuação de Propensão , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...