Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Genet ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801462

ABSTRACT

Granzyme B (GZMB), a critical member of the Gr gene family, is known to play an essential role in diverse physiological and pathological processes such as inflammation, acute and chronic inflammatory diseases, and cancer progression. In this study, we delve deeper into the role of GZMB within the context of gastric cancer (GC) to examine its expression patterns and functional implications. To accomplish this, we applied a combination of quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry techniques. These methodologies allowed us to accurately gauge GZMB expression levels in GC tissues and investigate their correlation with various clinical-pathological variables. Our secondary focus was to discern the regulatory influence of GZMB on GC cell biology. We used an array of assays including cell counting kit-8 (CCK-8), colony formation, 5-ethynyl-2'-deoxyuridine, and migration assays. The effect of GZMB on gastric cancer progression was further validated through a subcutaneous xenograft mouse model. Our findings underscored that GZMB mRNA and protein levels were upregulated in GC tissues, a feature that showed a significant correlation with GC staging. We also discovered that a decrease in GZMB expression via knockdown experiments suppressed the proliferation and migration capabilities of GC cells. This effect was manifested through diminished expression levels of epithelial-mesenchymal transition (EMT) markers. In stark contrast, the overexpression of GZMB through plasmid transfection appeared to enhance the proliferation and migration abilities of GC cells. This was coupled with an upregulation in EMT expression. Our study concludes by emphasizing that GZMB promotes the growth, migration, and EMT processes in gastric cancer. In vitro, cell-based experiments and in vivo xenograft mouse models confirm this. Our findings provide a more comprehensive understanding of GZMB's role in gastric cancer pathogenesis, potentially opening doors for novel therapeutic strategies targeting this molecular pathway.

2.
Adv Healthc Mater ; 13(6): e2303031, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37848188

ABSTRACT

Thermal ablation is a crucial therapeutic modality for hepatocellular carcinoma (HCC), but its efficacy is often hindered by the high recurrence rate attributed to insufficient ablation. Furthermore, the residual tumors following insufficient ablation exhibit a more pronounced immunosuppressive state, which accelerates the disease progression and leads to immune checkpoint blockade (ICB) resistance. Herein, evidence is presented that heightened intratumoral lactate accumulation, stemming from the augmented glycolytic activity of postablative residual HCC cells, may serve as a crucial driving force in exacerbating the immunosuppressive state of the tumor microenvironment (TME). To address this, an injectable nanoparticles-hydrogel composite system (LOX-MnO2 @Gel) is designed that gradually releases lactate oxidase (LOX)-loaded hollow mesoporous MnO2 nanoparticles at the tumor site to continuously deplete intratumoral lactate via a cascade catalytic reaction. Using subcutaneous and orthotopic HCC tumor-bearing mouse models, it is confirmed that LOX-MnO2 @Gel-mediated local lactate depletion can transform the immunosuppressive postablative TME into an immunocompetent one and synergizes with ICB therapy to significantly inhibit residual HCC growth and lung metastasis, thereby prolonging the survival of mice postablation. The work proposes an appealing strategy for synergistically combining antitumor metabolic therapy with immunotherapy to combat postablative HCC recurrence.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Animals , Mice , Lactic Acid , Carcinoma, Hepatocellular/therapy , Hydrogels , Manganese Compounds/pharmacology , Liver Neoplasms/therapy , Oxides , Immunotherapy , Tumor Microenvironment
3.
J Transl Med ; 21(1): 474, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37461041

ABSTRACT

BACKGROUND: The glycosyltransferase CHSY3 is a CHSY family member, yet its importance in the context of gastric cancer development remains incompletely understood. The present study was thus developed to explore the mechanistic importance of CHSY3 as a regulator of gastric cancer. METHODS: Expression of CHSY3 was verified by TCGA, GEO and HPA databases. Kaplan-Meier curve, ROC, univariate cox, multivariate cox, and nomogram models were used to verify the prognostic impact and predictive value of CHSY3. KEGG and GO methods were used to identify signaling pathways associated with CHSY3. TIDE and IPS scores were used to assess the immunotherapeutic value of CHSY3. WGCNA, Cytoscape constructs PPI networks and random forest models to identify key Hub genes. Finally, qRT-PCR and immunohistochemical staining were performed to verify CHSY3 expression in clinical specimens. The ability of CHSY3 to regulate tumor was further assessed by CCK-8 assay and cloning assay, EDU assay, migration assay, invasion assay, and xenograft tumor model analysis. RESULTS: The expression of CHSY3 was discovered to be abnormally upregulated in GC tissues through TCGA, GEO, and HPA databases, and the expression of CHSY3 was associated with poor prognosis in GC patients. Correlation analysis and Cox regression analysis revealed higher CHSY3 expression in higher T staging, an independent prognostic factor for GC. Moreover, elevated expression of CHSY3 was found to reduce the benefit of immunotherapy as assessed by the TIDE score and IPS score. Then, utilizing WGCNA, the PPI network constructed by Cytoscape, and random forest model, the Hub genes of COL5A2, POSTN, COL1A1, and FN1 associated with immunity were screened. Finally, the expression of CHSY3 in GC tissues was verified by qRT-PCR and immunohistochemical staining. Moreover, the expression of CHSY3 was further demonstrated by in vivo and in vitro experiments to promote the proliferation, migration, and invasive ability of GC. CONCLUSIONS: The results of this study suggest that CHSY3 is an important regulator of gastric cancer progression, highlighting its promise as a therapeutic target for gastric cancer.


Subject(s)
Stomach Neoplasms , Animals , Humans , Biological Assay , Cell Proliferation/genetics , Databases, Factual , Disease Models, Animal , Stomach Neoplasms/genetics
4.
Tissue Cell ; 82: 102119, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37257286

ABSTRACT

Dishevelled family proteins (DVL1-3), key scaffold proteins, act on canonical and non-canonical Wnt/ß-catenin signaling pathway. DVL has been implicated in various tumor progression. However, its role and underlying mechanisms in gastric cancer (GC) remain unclear. The aim of this study was to investigate the role of DVL in GC development using cell lines and 209 GC specimens. We analyzed three orthologs of DVL in GC tissues and paired adjacent non-tumor tissues, and only DVL2 is highly expressed in GC tissues. We also analyzed clinicopathological data on DVL2 expression in gastric cancer specimens. In immunohistochemistry, DVL2 expression was up-regulated in GC tissues compared with paired adjacent non-tumor tissues (153/209, 73.2%). DVL2 expression level was significantly correlated with many clinicopathological parameters such as T stage (P < 0.001) and N stage (P < 0.001). Survival analysis showed that the overall survival (OS) of patients with high expression of DVL2 was significantly shorter than those with low expression. Multivariate Cox regression analysis revealed that DVL2 expression was an important and independent prognostic factor for gastric cancer patients (P = 0.011, HR=1.78, 95%CI (1.14-2.79). Depletion of endogenous DVL2 using short hairpin RNA (shRNA) inhibited GC cell proliferation, migration, and invasion. The abnormal activation of Wnt/ß-catenin signaling pathway is mainly achieved through the abnormal expression of DVL2. DVL2 is highly expressed in gastric cancer tissues, which may be a new independent risk factor for the prognosis of gastric cancer patients. In gastric cancer, DVL2 overexpression plays a crucial role in the occurrence and development of gastric cancer, so it may become a new, effective and complementary therapeutic target for gastric cancer.


Subject(s)
Stomach Neoplasms , Wnt Signaling Pathway , Humans , Wnt Signaling Pathway/genetics , Stomach Neoplasms/genetics , beta Catenin/metabolism , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Cell Line , RNA, Small Interfering , Cell Line, Tumor , Cell Proliferation/genetics
5.
Cancer Med ; 12(1): 837-851, 2023 01.
Article in English | MEDLINE | ID: mdl-35634680

ABSTRACT

BACKGROUND: PI4K2A has been found to have a tumor-promoting role in various solid tumors and be involved in various biological procedures. In this article, we aim to investigate the prognostic values of PI4K2A and provide new insights in colon adenocarcinoma (COAD). METHODS: The Cancer Genome Atlas (TCGA) database, Human Protein Atlas online database, and UALCAN database were used to analyze the expression of PI4K2A in COAD and the survival of patients. Univariate and multifactorial Cox regression analyses were used to assess the prognosis of PI4K2A on COAD. GSEA was used to explore PI4K2A-related signaling pathways. In addition, the effect of PI4K2A on immune checkpoint inhibitors (ICIs) treatment was investigated by constructing a TIDE model and predicting the association between PI4K2A and anticancer drug sensitivity through the CellMiner database. RESULTS: In the TCGA database, PI4K2A was highly expressed in COAD and the similar results were verified by qRT-PCR. Survival analysis, utilizing Kaplan-Meier curves, revealed that COAD patients with high PI4K2A expression had a worse prognosis. In addition, PI4K2A expression was discovered to have been associated with T-stage, N-stage, and pathological stage by logistic analysis. Next, we utilized univariate and multifactorial Cox regression analyses to identify PI4K2A as an independent predictor. Additionally, GSEA analysis indicates that PI4K2A is enriched in MAPK signaling pathway, Toll-like receptor signaling pathway, etc. In COAD, PI4K2A was remarkably associated with the tumor immune microenvironment. In addition, by constructing a TIDE model, we discovered that COAD patients in the PI4K2A low-expression cohort were better treated with ICI. Finally, analysis of the CellMiner database predicted that PI4K2A was adversely correlated with the sensitivity of various anticancer drugs. CONCLUSIONS: Our study suggests that PI4K2A may be a potential predictor of poor prognosis in COAD and a potential biomarker for early diagnosis, prognosis, and treatment.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Humans , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Databases, Factual , Immune Checkpoint Inhibitors , MAP Kinase Signaling System , Prognosis , Tumor Microenvironment
6.
Sci Rep ; 12(1): 21282, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36482181

ABSTRACT

N7-methyladenosine (m7G) modifications have been the subject of growing research interest with respect to their relationship with the progression and treatment of various cancers. This analysis was designed to examine the association between m7G-related gene expression and colorectal cancer (CRC) patient outcomes. Initial training analyses were performed using the TCGA dataset, with the GSE28722 dataset then being used to validate these results. Univariate Cox analyses were initially conducted to screen out prognostic m7G-related genes, after which a LASSO approach was used to construct an m7G risk score (MRS) model. Kaplan-Meier curves, ROC curves, and Cox analyses were subsequently used to validate the prognostic utility of this model in CRC patients. The R maftools package was further employed to assess mutational characteristics in CRC patients in different MRS subgroups, while the ESTIMATE, CIBERSORT, and ssGSEA tools were used to conduct immune infiltration analyses. A WGCNA was then performed to identify key immune-associated hub genes. The EIF4E3, GEMIN5, and NCBP2 genes were used to establish the MRS model. Patients with high MRS scores exhibited worse overall survival than patients with low scores. In Cox analyses, MRS scores were independently associated with CRC patient prognosis. Patients with low MRS scores exhibited a higher tumor mutational burden and higher levels of microsatellite instability. In immune infiltration analyses, higher immune checkpoint expression and greater immune cell infiltration were also observed in patients with low MRS scores. WGCNA analyses further identified 25 CD8+ T cell infiltration-associated genes. These findings suggest that MRS values represent a useful biomarker capable of differentiating among CRC patients with different immunological features and prognostic outcomes, offering an opportunity to better determine which patients are likely to benefit from immune checkpoint inhibitor treatment.


Subject(s)
Immune Checkpoint Inhibitors , Microsatellite Instability , Humans , Prognosis , ROC Curve , Risk Factors
7.
BMC Urol ; 18(1): 33, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29739380

ABSTRACT

BACKGROUND: To observe the efficacy of self-help position therapy (SHPT) after holmium laser lithotripsy via flexible ureteroscopy (FURS). METHODS: From January 2010 to November 2015, 736 nephrolithiasis patients who had received FURS lithotripsy were analyzed retrospectively. In position group, 220 cases accepted SHPT after lithotripsies, and 428 cases as control, coming from another independent inpatient area in the same center. The stone-free status (SFS) between two groups were compared at the 2nd, 4th and 12th week ends by X-ray examinations. RESULTS: The preoperative incidence of hydronephrosis (25.9% vs. 18.0%, p = 0.018) or lower calyceal seeper (33.6% vs. 24.3%, p = 0.012) and the proportion of patients with > 2.0 cm stones (33.6% vs. 24.3%, p = 0.003) were all significantly higher in position group than in control group. There were no substantial difference between two groups in age, BMI, gender and medical histories. In postoperative followup, the incidence of hydronephrosis in position group was significantly lower than in control group (9.5% vs. 15.7%, p = 0.032) after removing double-J stents. In position group, the SFS of the 2nd week end (60.9% vs. 47.2%, p = 0.001), the 4th week end (74.1% vs. 62.8%, p = 0.004) and the 12th week end (86.9% vs. 79.4%, p = 0.021) were all significantly higher than those in control group. CONCLUSIONS: SHPT after holmium laser lithotripsy via FURS may increase postoperative SFS, accelerate stone fragment clearance, and decrease the incidence of hydronephrosis after removal of double-J stents. The therapy does not require professional assistance and is economical, simple, and effective.


Subject(s)
Holmium , Lithotripsy, Laser/methods , Nephrolithiasis/therapy , Patient Positioning/methods , Self Care/methods , Ureteroscopy/methods , Adult , Female , Follow-Up Studies , Humans , Lithotripsy, Laser/instrumentation , Male , Middle Aged , Nephrolithiasis/diagnostic imaging , Retrospective Studies , Treatment Outcome , Ureteroscopes/statistics & numerical data , Ureteroscopy/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...