Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.463
Filter
1.
Phytomedicine ; 134: 155984, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39265444

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) are involved in the progression of gastric cancer (GC) as a critical component of the tumor microenvironment (TME), yet specific interventions remain limited. Natural products hold a promising application prospect in the field of anti-tumor in view of their high activity and ease of binding with biological macromolecules. However, the role of natural products in modulating the cross-talk between CAFs and GC cells has not been fully investigated. PURPOSE: The aim of this study was to identify a potential therapeutic target in CAFs and then screen for natural small molecule drugs with anti-tumor activity against this target. METHODS: Integrating bioinformatics analysis of public databases and experimental validation of human samples and cell lines to identify a candidate target in CAFs. Molecular docking and biolayer interferometry technique were utilized for screening potential natural small molecule drugs. The efficacy and underlying mechanisms of the candidates were explored in vitro and in vivo through techniques such as lentiviral infection, cell spheroids culture, immunoprecipitation and cells-derived xenografts. RESULTS: IL18 receptor accessory protein (IL18RAP) was found to be overexpressed in CAFs derived from GC tissues and facilitated the protumor function of CAFs on GC. Based on virtual screening and experimental validation, we identified a natural product, eupafolin, that interfered with IL18 signaling. Phenotyping studies confirmed that the proliferation, spheroids formation and tumorigenesis of GC cells facilitated by CAFs were greatly attenuated by eupafolin both in vitro and in vivo. Mechanistically, eupafolin impeded the formation of IL18 receptor (IL18R) complex by directly binding to IL18RAP, thus blocking IL18-mediated nuclear factor kappa B (NF-κB) activation and reduced the synthesis and secretion of IL6 in CAFs. As a consequence, it inactivated signal transducer and activator of transcription 3 (STAT3) in GC cells. CONCLUSION: This study provides new evidence that IL18 signaling regulates the cross-talk between GC cells and CAFs. And it highlights a novel pharmacological role of eupafolin in inhibiting IL18 signaling, thereby curbing the development of GC via modulating CAFs.


Subject(s)
Cancer-Associated Fibroblasts , Interleukin-18 , Signal Transduction , Stomach Neoplasms , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Humans , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Interleukin-18/metabolism , Mice, Nude , Molecular Docking Simulation , Mice , Tumor Microenvironment/drug effects , Mice, Inbred BALB C
3.
J Cancer Res Clin Oncol ; 150(9): 416, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249161

ABSTRACT

BACKGROUND: Gastric cancer (GC), a prevalent malignant tumor which is a leading cause of death from malignancy around the world. Peritoneal metastasis accounts for the major cause of mortality in patients with GC. Despite hyperthermia intraperitoneal chemotherapy (HIPEC) improves the therapeutic effect of GC, it's equivocal about the mechanism under HIPEC. METHODS: MiR-183-5p expression was sifted from miRNA chip and detected in both GC patients and cell lines by qRT-PCR. Gene interference and rescue experiments were performed to identified biological function in vitro and vivo. Next, we affirmed PPP2CA as targeted of miR-183-5p by dual luciferase reporter assay. Finally, the potential relationship between HIPEC and miR-183-5p was explored. RESULTS: MiR-183-5p is up-regulated in GC and associated with advanced stage and poor prognosis. MiR-183-5p accelerate GC migration in vitro which is influenced by miR-183-5p/PPP2CA/AKT/GSK3ß/ß-catenin Axis. HIPEC exerts migration inhibition via attenuating miR-183-5p expression. CONCLUSION: MiR-183-5p can be used as a potential HIPEC biomarker in patients with CC.


Subject(s)
Cell Movement , Glycogen Synthase Kinase 3 beta , Hyperthermia, Induced , MicroRNAs , Proto-Oncogene Proteins c-akt , Stomach Neoplasms , beta Catenin , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , MicroRNAs/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Hyperthermia, Induced/methods , beta Catenin/metabolism , beta Catenin/genetics , Proto-Oncogene Proteins c-akt/metabolism , Mice , Animals , Male , Female , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Cell Line, Tumor , Mice, Nude , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic , Prognosis , Middle Aged , Mice, Inbred BALB C , Antineoplastic Agents, Phytogenic/pharmacology
4.
BMC Cancer ; 24(1): 1117, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251966

ABSTRACT

BACKGROUND/AIMS: Gastric cancer (GC) ranks among the prevalent types of cancer, and its progression is influenced by the tumor microenvironment (TME). A comprehensive comprehension of the TME associated with GC has the potential to unveil therapeutic targets of significance. METHODS: The complexity and heterogeneity of TME interactions were revealed through our investigation using an integrated analysis of single-cell and bulk-tissue sequencing data. RESULTS: We constructed a single-cell transcriptomic atlas of 150,913 cells isolated from GC patients. Our analysis revealed the intricate nature and heterogeneity of the GC TME and the metabolic properties of major cell types. Furthermore, two cell subtypes, LOX+ Fibroblasts and M2 Macrophages, were enriched in tumor tissue and related to the outcome of GC patients. In addition, LOX+ Fibroblasts were significantly associated with M2 macrophages. immunofluorescence double labeling indicated LOX+ Fibroblasts and M2 Macrophages were tightly localized in GC tissue. The two cell subpopulations strongly interacted in a hypoxic microenvironment, yielding an immunosuppressive phenotype. Our findings further suggest that LOX+ Fibroblasts may act as a trigger for inducing the differentiation of monocytes into M2 Macrophages via the IL6-IL6R signaling pathway. CONCLUSIONS: Our study revealed the intricate and interdependent communication network between the fibroblast and macrophage subpopulations, which could offer valuable insights for targeted manipulation of the tumor microenvironment.


Subject(s)
Fibroblasts , Macrophages , Single-Cell Analysis , Stomach Neoplasms , Tumor Microenvironment , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Tumor Microenvironment/immunology , Single-Cell Analysis/methods , Macrophages/metabolism , Macrophages/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Scavenger Receptors, Class E/metabolism , Scavenger Receptors, Class E/genetics , Cell Communication/immunology , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Transcriptome , Signal Transduction
5.
Cancer Control ; 31: 10732748241281716, 2024.
Article in English | MEDLINE | ID: mdl-39236066

ABSTRACT

INTRODUCTION: The role of SMU1 in DNA replication and RNA splicing is well-established, yet its specific function and dysregulated mechanisms in gastric cancer (GC) remain inadequately elucidated. This study seeks to investigate the potential oncogenic and progression-promoting effects of SMU1 in GC, with the ultimate goal of informing novel approaches for treatment and diagnosis. METHODS: The study investigated the expression levels of SMU1 in GC and adjacent normal tissues by analyzing data from the TCGA (27 tissue pairs) and GEO (47 tissue pairs) databases. Immunohistochemistry was used to examine 277 tumor tissue and adjacent non-tumor tissue spots from GC tissue chips, along with relevant follow-up information. The study further assessed the proliferation, invasion, and migration capabilities of cells by manipulating SMU1 expression levels and conducting various assays, including CCK-8, EdU incorporation, colony formation, transwells, flow cytometry, and subcutaneous tumorigenesis assays. RESULTS: Our study revealed a significant upregulation of SMU1 mRNA and protein levels in GC tissues compared to adjacent tissues. Univariate and multivariate Cox analysis demonstrated that elevated levels of SMU1 were independent prognostic factors for GC prognosis (P = 0.036). Additionally, median survival analysis indicated a significant association between high SMU1 expression and poor prognosis in GC patients (P = 0.0002). In experiments conducted both in vivo and in vitro, it was determined that elevated levels of SMU1 can enhance the proliferation, invasion, and migration of GC cells, whereas suppression of SMU1 can impede the progression of GC by modulating the G1/S checkpoint of the cell cycle. CONCLUSIONS: Our research introduces the novel idea that SMU1 could serve as a prognostic marker for GC progression, influencing cell proliferation through cell cycle activation. These results offer valuable insights into the understanding, diagnosis, and management of gastric carcinoma.


Subject(s)
Cell Cycle , Cell Movement , Cell Proliferation , Neoplasm Invasiveness , Stomach Neoplasms , Animals , Female , Humans , Male , Mice , Middle Aged , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Mice, Nude , Neoplasm Invasiveness/genetics , Prognosis , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
6.
PLoS One ; 19(9): e0309014, 2024.
Article in English | MEDLINE | ID: mdl-39241034

ABSTRACT

5-Fluorouracil (5-FU) is widely used in the treatment of gastric cancer, and the emergence of drug resistance and toxic effects has limited its application. Therefore, there is an urgent need for safe and effective novel drugs or new therapies. ß-Ionone (BI) is found in vegetables and fruits and possesses an inhibitory proliferation of tumor cells in vitro and in vivo. In this study, we investigated whether BI could enhance the inhibitory effects of 5-FU on the proliferation of gastric adenocarcinoma cells and the growth of gastric cancer cell xenografts in a mouse model. The effects of BI and 5-FU alone or their combination on the cell viability, apoptosis, and mitochondrial membrane potential, the cell cycle, and its related proteins-Cyclin D1, and CDK4 as well as PCNA and GSK-3ß were evaluated in SGC-7901 cells and MKN45 cells by MTT, MB, flow cytometry and Western blot. In addition, the effects of BI and 5-FU alone or their combination on the growth of SGC-7901 cell xenografts in nude mice were investigated. The results showed that BI significantly enhanced the sensitivity of gastric adenocarcinoma cells to 5-FU in vitro and in vivo, i.e. proliferation inhibited, apoptosis induced and GSK-3ß protein activated. Therefore, our results suggest that BI increases the antitumor effect of 5-FU on gastric adenocarcinoma cells, at least partly from an activated GSK-3ß signaling pathway.


Subject(s)
Adenocarcinoma , Apoptosis , Cell Proliferation , Fluorouracil , Glycogen Synthase Kinase 3 beta , Mice, Nude , Norisoprenoids , Signal Transduction , Stomach Neoplasms , Animals , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Fluorouracil/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Cell Proliferation/drug effects , Signal Transduction/drug effects , Cell Line, Tumor , Norisoprenoids/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Mice , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Drug Synergism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Glycogen Synthase Kinase 3/metabolism , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/metabolism
7.
Funct Integr Genomics ; 24(5): 175, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39325107

ABSTRACT

Gastric cancer (GC) remains a major public health challenge worldwide. Long non-coding RNAs (lncRNAs) play important roles in the development, progression, and resistance to the treatment of GC, as shown by recent developments in molecular characterization. Still, an in-depth investigation of the lncRNA landscape in GC is absent. However, The objective of this systematic review is to evaluate our present understanding of the role that lncRNA dysregulation plays in the etiology of GC and treatment resistance, with a focus on the underlying mechanisms and clinical implications. Research that described the functions of lncRNA in angiogenesis, stemness, epigenetics, metastasis, apoptosis, development, and resistance to key treatments was given priority. In GC, it has been discovered that a large number of lncRNAs, including MALAT1, HOTAIR, H19, and ANRIL, are aberrantly expressed and are connected with disease-related outcomes. Through various methods such as chromatin remodeling, signal transduction pathways, and microRNA sponging, they modulate hallmark cancer capabilities. Through the activation of stemness programs, epithelial-mesenchymal transition (EMT), and survival signaling, LncRNAs also control resistance to immunotherapy, chemotherapy, and targeted therapies. By clarifying their molecular roles further, we may be able to identify new treatment targets and ways to overcome resistance. This article aims to explore the interplay between lncRNAs, and GC. Specifically, the focus is on understanding how lncRNAs contribute to the etiology of GC and influence treatment resistance in patients with this disease.


Subject(s)
Drug Resistance, Neoplasm , RNA, Long Noncoding , Stomach Neoplasms , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Humans , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic
8.
Cell Death Dis ; 15(9): 670, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266534

ABSTRACT

Cancer cells autonomously alter metabolic pathways in response to dynamic nutrient conditions in the microenvironment to maintain cell survival and proliferation. A better understanding of these adaptive alterations may reveal the vulnerabilities of cancer cells. Here, we demonstrate that coactivator-associated arginine methyltransferase 1 (CARM1) is frequently overexpressed in gastric cancer and predicts poor prognosis of patients with this cancer. Gastric cancer cells sense a reduced extracellular glucose content, leading to activation of nuclear factor erythroid 2-related factor 2 (NRF2). Subsequently, NRF2 mediates the classic antioxidant pathway to eliminate the accumulation of reactive oxygen species induced by low glucose. We found that NRF2 binds to the CARM1 promoter, upregulating its expression and triggering CARM1-mediated hypermethylation of histone H3 methylated at R arginine 17 (H3R17me2) in the glucose-6-phosphate dehydrogenase gene body. The upregulation of this dehydrogenase, driven by the H3R17me2 modification, redirects glucose carbon flux toward the pentose phosphate pathway. This redirection contributes to nucleotide synthesis (yielding nucleotide precursors, such as ribose-5-phosphate) and redox homeostasis and ultimately facilitates cancer cell survival and growth. NRF2 or CARM1 knockdown results in decreased H3R17me2a accompanied by the reduction of glucose-6-phosphate dehydrogenase under low glucose conditions. Collectively, this study reveals a significant role of CARM1 in regulating the tumor metabolic switch and identifies CARM1 as a potential therapeutic target for gastric cancer treatment.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glucose , NF-E2-Related Factor 2 , Pentose Phosphate Pathway , Protein-Arginine N-Methyltransferases , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Pentose Phosphate Pathway/genetics , Glucose/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Cell Line, Tumor , Animals , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/genetics , Mice , Reactive Oxygen Species/metabolism , Histones/metabolism , Promoter Regions, Genetic/genetics , Mice, Nude , Transcription, Genetic , Cell Proliferation/genetics
9.
J Transl Med ; 22(1): 832, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256760

ABSTRACT

BACKGROUND: The roles of the transcriptional factor SIX2 have been identified in several tumors. However, its roles in gastric cancer (GC) progression have not yet been revealed. Our objective is to explore the impact and underlying mechanisms of SIX2 on the stemness of GC cells. METHODS: Lentivirus infection was employed to establish stable expression SIX2 or PFN2 in GC cells. Gain- and loss-of-function experiments were conducted to detect changes of stemness markers, flow cytometry profiles, tumor spheroid formation, and tumor-initiating ability. ChIP, RNA-sequencing, tissue microarray, and bioinformatics analysis were performed to reveal the correlation between SIX2 and PFN2. The mechanisms underlying the SIX2/PFN2 loop-mediated effects were elucidated through tissue microarray analysis, RNA stability assay, IP-MS, Co-Immunoprecipitation, and inhibition of the JNK signaling pathway. RESULTS: The stemness of GC cells was enhanced by SIX2. Mechanistically, SIX2 directly bound to PFN2's promoter and promoted PFN2 activity. PFN2, in turn, promoted the mRNA stability of SIX2 by recruiting RNA binding protein YBX-1, subsequently activating the downstream MAPK/JNK pathway. CONCLUSION: This study unveils the roles of SIX2 in governing GC cell stemness, defining a novel SIX2/PFN2 regulatory loop responsible for this regulation. This suggests the potential of targeting the SIX2/PFN2 loop for GC treatment (Graphical Abstracts).


Subject(s)
Feedback, Physiological , Gene Expression Regulation, Neoplastic , Homeodomain Proteins , Neoplastic Stem Cells , Profilins , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Humans , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Profilins/metabolism , Profilins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Animals , Promoter Regions, Genetic/genetics , RNA Stability/genetics , MAP Kinase Signaling System , Protein Binding
10.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273151

ABSTRACT

Gastric cancer is one of the most common cancers worldwide, and new therapeutic strategies are urgently needed. Ferroptosis is an intracellular iron-dependent cell death induced by the accumulation of lipid peroxidation, a mechanism different from conventional apoptosis and necrosis. Therefore, induction of ferroptosis is expected to be a new therapeutic strategy. Glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1) have been identified as the major inhibitors of ferroptosis. Herein, we performed immunohistochemistry for GPX4, FSP1, and 4-HNE using tissues from patients with gastric cancer and investigated the relationship between these factors and prognosis. Patients with high GPX4 expression or high GPX4 expression and low 4-HNE accumulation tended to have a poor prognosis (p = 0.036, 0.023), whereas those with low FSP1 expression and high 4-HNE accumulation had a good prognosis (p = 0.033). The synergistic induction of cell death by inhibiting GPX4 and FSP1 in vitro was also observed, indicating that the cell death was non-apoptotic. Our results indicate that the expression and accumulation of lipid peroxidation-related factors play an important role in the clinicopathological significance of gastric cancer and that novel therapeutic strategies targeting GPX4 and FSP1 may be effective in treating patients with gastric cancer who have poor prognosis.


Subject(s)
Biomarkers, Tumor , Ferroptosis , Lipid Peroxidation , Phospholipid Hydroperoxide Glutathione Peroxidase , Stomach Neoplasms , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Prognosis , Female , Male , Biomarkers, Tumor/metabolism , Aged , Middle Aged , Ferroptosis/drug effects , Cell Line, Tumor , Aldehydes/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics
11.
Sci Signal ; 17(854): eabq4888, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39288218

ABSTRACT

Doublecortin-like kinase 1 (DCLK1) is a proposed driver of gastric cancer (GC) that phosphorylates serine and threonine residues. Here, we showed that the kinase activity of DCLK1 orchestrated cancer cell-intrinsic and-extrinsic processes that led to pro-invasive and pro-metastatic reprogramming of GC cells. Inhibition of the kinase activity of DCLK1 reduced the growth of subcutaneous xenograft tumors formed from MKN1 human gastric carcinoma cells in mice and decreased the abundance of the stromal markers α-Sma, vimentin, and collagen. Similar effects were seen in mice with xenograft tumors formed from MKN1 cells expressing a kinase-inactive DCLK1 mutant (MKN1D511N). MKN1D511N cells also had reduced in vitro migratory potential and stemness compared with control cells. Mice orthotopically grafted with MKN1 cells overexpressing DCLK1 (MKN1DCLK1) showed increased invasiveness and had a greater incidence of lung metastases compared with those grafted with control MKN1 cells. Mechanistically, we showed that the chemokine CXCL12 acted downstream of DCLK1 in cultured MKN1 cells and in mice subcutaneously implanted with gastric tumors formed by MKN1DCLK1 cells. Moreover, inhibition of the kinase activity of DCLK1 or the expression of DCLK1D511N reversed the pro-tumorigenic and pro-metastatic phenotype. Together, this study establishes DCLK1 as a broadly acting and potentially targetable promoter of GC.


Subject(s)
Disease Progression , Doublecortin-Like Kinases , Intracellular Signaling Peptides and Proteins , Phenotype , Protein Serine-Threonine Kinases , Stomach Neoplasms , Doublecortin-Like Kinases/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Animals , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Cell Line, Tumor , Cell Movement/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism
12.
Sci Rep ; 14(1): 21648, 2024 09 17.
Article in English | MEDLINE | ID: mdl-39289452

ABSTRACT

Helicobacter pylori (H. pylori) is one of the most common bacterial infections in the world, and its key virulence component CagA is the leading cause of gastric cancer. Mitophagy is a form of selective autophagy that eliminates damaged mitochondria and is essential for some viruses and bacteria to evade the immune system. However, the mechanisms by which CagA mediates H. pylori-induced mitophagy and NLRP3 inflammasome activation remain elusive. In this study, we reported that H. pylori primarily uses its CagA to induce mitochondrial oxidative damage, mitochondrial dysfunction, dynamic imbalance, and to block autophagic flux. Inhibition of mitophagy led to an increase in NLRP3 inflammasome activation and apoptosis and a decrease in the viability of H. pylori-infected cells. Our findings suggested that H. pylori induces mitochondrial dysfunction and mitophagy primarily via CagA. It reduces NLRP3 inflammasome activation to evade host immune surveillance and increases the survival and viability of infected cells, potentially leading to gastric cancer initiation and development. Our findings provide new insights into the pathogenesis of H. pylori-induced gastric cancer, and inhibition of mitophagy may be one of the novel techniques for the prevention and treatment of this disease.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Helicobacter pylori , Inflammasomes , Mitochondria , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Inflammasomes/metabolism , Helicobacter pylori/pathogenicity , Helicobacter pylori/physiology , Humans , Mitochondria/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/immunology , Cell Survival , Stomach Neoplasms/microbiology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Apoptosis
13.
Clinics (Sao Paulo) ; 79: 100492, 2024.
Article in English | MEDLINE | ID: mdl-39293372

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) hold critical importance due to their notable function in developing Gastric Cancer (GC), which is a malignancy with the third most frequent occurrence worldwide. The aim of this study was to see if circRNA_0044516 would control GC cell proliferation and establish more effective therapeutic strategies. METHODS: In GC tissues or cells, quantitative Real­Time Polymerase Chain Reaction (qRT-PCR) was employed for the detection of the expression of circRNA_100349, Insulin-like Growth Factor II (IGF2), and miR-218-5p. CCK-8 assays were employed to gauge the proliferation of cells. A luciferase reporter was employed to establish the relationship of circRNA_100349 or IGF2 with miR-218-5p. RESULTS: CircRNA_100349 was observed to undergo upregulation in GC cell lines along with tissues. GC cell proliferation was prevented by downregulating circRNA_100349. MiR-149 was targeted by CircRNA_100349, and its downregulation increased the amount of miR-218-5p in GC cells. Simultaneously silencing circRNA_100349 decreased IGF2 expression via miR-218-5p, and thus suppressed GC cell proliferation. Furthermore, in nude mice, circRNA_100349 knockdown prevented the tumor development of GC cells. CONCLUSIONS: The findings furnished evidence of the critical involvement of circRNA_100349 in GC and that its downregulation impedes GC cell proliferation via the miR-218-5p/IGF2 axis.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II , MicroRNAs , RNA, Circular , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Cell Proliferation/genetics , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Animals , Down-Regulation , Up-Regulation , Mice, Nude , Mice , Real-Time Polymerase Chain Reaction , Male
14.
Int J Biol Sci ; 20(12): 4978-4998, 2024.
Article in English | MEDLINE | ID: mdl-39309429

ABSTRACT

Gastric cancer (GC) ranks as the fifth most common cancer and the fourth leading cause of cancer-related deaths globally. Despite advancements in molecular profiling, the mechanisms driving GC proliferation and metastasis remain unclear. This study identifies Early 2 Factor 4 (E2F4) as a key transcription factor that promotes GC cell proliferation, migration, and invasion by upregulating DNA Replication and Sister Chromatid Cohesion 1 (DSCC1) expression. Bioinformatics and transcription factor analyses revealed E2F4 as a significant regulator of DSCC1. Functional assays confirmed E2F4's role in enhancing GC cell malignancy in vitro and in vivo. Knockdown and overexpression experiments demonstrated that E2F4 positively regulates DSCC1 at the transcriptional level, with ChIP-qPCR and dual luciferase reporter assays validating the binding sites on the DSCC1 promoter. These findings highlight the E2F4-DSCC1 axis as a potential therapeutic target to mitigate GC progression.


Subject(s)
Cell Movement , Cell Proliferation , E2F4 Transcription Factor , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Humans , Cell Proliferation/genetics , Cell Movement/genetics , Cell Line, Tumor , E2F4 Transcription Factor/metabolism , E2F4 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Animals , Mice , Mice, Nude , Neoplasm Invasiveness
15.
Int J Biol Sci ; 20(12): 4819-4837, 2024.
Article in English | MEDLINE | ID: mdl-39309430

ABSTRACT

Gastric cancer (GC) poses global challenges due to its difficult early diagnosis and drug resistance, necessitating the identification of early detection markers and understanding of oncogenic pathways for effective GC therapy. Endothelial cell-specific molecule 1 (ESM1), a secreted glycoprotein, is elevated in various cancers, but its role in GC remains controversial. In our study, ESM1 was elevated in GC tissues, and its concentration was correlated with progression and poorer patient prognosis in independent cohorts. Functionally, ESM1 expression promoted proliferation, anoikis resistance, and motility of GC cells, as well as tumor growth in PDOs and in GC xenograft models. Mechanistically, ESM1 expression triggered the epithelial-to-mesenchymal transition (EMT) of GC cells by enhancing epidermal growth factor receptor (EGFR)/human EGFR 3 (HER3) association and activating the EGFR/HER3-Akt pathway. Additionally, angiopoietin-2 (ANGPT2) was found to be highly correlated with ESM1 and interplayed with Akt to induce the EMT and cancer progression. Use of a signal peptide deletion mutant (ESM1-19del) showed that the secreted form of ESM1 is crucial for its protumorigenic effects by activating the EGFR/HER3-Akt/ANGPT2 pathway to promote the EMT. Patients with high levels of both ESM1 and ANGPT2 had the poorest prognoses. Furthermore, therapeutic peptides successfully inhibited ESM1's induction of the aforementioned signals and motility of GC cells. ESM1's oncogenic role in GC involves activating the EGFR/HER3-Akt/ANGPT2 pathway, presenting a potential therapeutic target for GC.


Subject(s)
Angiopoietin-2 , Epithelial-Mesenchymal Transition , ErbB Receptors , Proteoglycans , Proto-Oncogene Proteins c-akt , Signal Transduction , Stomach Neoplasms , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Humans , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Proteoglycans/metabolism , Cell Line, Tumor , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Mice , Receptor, ErbB-3/metabolism , Male , Female , Cell Proliferation , Mice, Nude
16.
Immun Inflamm Dis ; 12(9): e70004, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39254476

ABSTRACT

INTRODUCTION: The purpose of this research was to determine how the P53/microRNA-34a (miR-34a)/survivin pathway contributes to oxaliplatin-induced (L-OHP) cell inhibition in gastric cancer. METHODS: The BGC-823 gastric cancer cells were selected, and we examined their viability following treatment with L-OHP at different concentrations and time periods. The expression levels of miR-34a, P53, and survivin in the cells were determined. RESULTS: In the 12- and 24-h groups, drug concentration of 15 µg/cm² (p < .005 in both) significantly lowered cell viability. In comparison to the control group, miR-34a mRNA expression, P53 mRNA expression, and protein expression were all significantly greater in the 24-h group (p = .0324, p = .0069, p = .0260, respectively), but survivin mRNA and protein expressions were significantly lower than those in the control group (p = .0338, p = .0032, respectively). There was a significant decrease in gastric cancer cells in the miR-34a overexpression group (p = .0020), a significant increase in P53 mRNA and protein expression compared to the control group (p = .0080, p = .0121, respectively), and a significant decrease in survivin mRNA and protein expression compared to the control group. (p = .0213, p = .0069, respectively). CONCLUSION: Oxaliplatin inhibits tumor growth, invasion, and metastasis by upregulating miR-34a, activating the expression of the upstream P53 gene, and driving the downregulation of survivin (P53/miR-34a/survivin axis) in BGC-823 gastric cancer cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Inhibitor of Apoptosis Proteins , MicroRNAs , Oxaliplatin , Stomach Neoplasms , Survivin , Tumor Suppressor Protein p53 , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , MicroRNAs/genetics , Humans , Oxaliplatin/pharmacology , Survivin/metabolism , Survivin/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Antineoplastic Agents/pharmacology , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Cell Survival/drug effects , Signal Transduction/drug effects , Cell Proliferation/drug effects , Disease Progression
17.
J Agric Food Chem ; 72(38): 20918-20929, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39262347

ABSTRACT

Royal jelly (RJ) is a natural food product with nutritional value and anticancer activity. However, their effects on gastric cancer are unclear. Here, we show that treatment with 5-320 µg/mL of RJ, ethanol extract (RJEE), and protein hydrolyzate (RJPH) decreased the viability of MKN-28 gastric cancer cells, with a half-maximal inhibitory concentration of 123.22 µg/mL for RJEE. RJ, RJEE, and RJPH increase the lactate dehydrogenase release rate and change the morphology of the cells, resulting in cell shrinkage, nucleoplasm condensation, and the formation of apoptotic bodies. RJ and its functional components stagnated the cell cycle in the G0/G1 phase, accompanied by the accumulation of reactive oxygen species, decreased mitochondrial membrane potential, and increased expression levels of p53 and p21 proteins, caspase-3 activation, and apoptosis. Therefore, RJ, RJEE, and RJPH have potential inhibitory effects on the proliferation of gastric cancer cells.


Subject(s)
Apoptosis , Cell Proliferation , Fatty Acids , Stomach Neoplasms , Humans , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Apoptosis/drug effects , Fatty Acids/chemistry , Fatty Acids/pharmacology , Fatty Acids/metabolism , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Caspase 3/genetics , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics
18.
Mol Biol Rep ; 51(1): 998, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39299971

ABSTRACT

BACKGROUND: Myricetin, a flavanol present in fruits, tea, and vegetables, has the potential to reduce chronic diseases like gastric cancer by promoting cell death and stopping cell growth. However, its limited bioactivity due to its short lifespan and poor solubility in water has been a challenge. The current research focuses on incorporating myricetin into alginate-cellulose hybrid nanocrystals to enhance its selective proapoptotic effects on human AGS gastric cancer cells. METHODS: MAC-NCs, myricetin-loaded alginate-cellulose hybrid nanocrystals, were synthesized using a combined co-precipitation/ultrasonic homogenization method and characterized through Dynamic Light Scattering (DLS), Fourier Transform Infrared Spectroscopy (FTIR), Field Emission Scanning Electron Microscope (FESEM), and Zeta-potential analyses. Their cytotoxic activity was tested on cancerous (AGS) and normal (Huvec) cells, revealing selective toxicity. Apoptotic markers, Caspase 8 and Caspase 9, gene expression was measured, and cell death type was confirmed using DAPI staining and flow cytometry on AGS cells. RESULTS: Synthesized MAC-NCs, measuring 40 nm, showed significant selective toxicity on human gastric cells (IC50 of 31.05 µg/mL) compared to normal endothelial cells (IC50 of 214.26 µg/mL). DAPI and annexin flow cytometry revealed increased apoptotic bodies in gastric cells, indicating apoptosis. However, the apoptosis was found to be independent of Caspase-8 and Caspase-9. CONCLUSION: The current study provides critical insights into the therapeutic potential of MAC-NCs for gastric cancer treatment. Based on the notable induction of apoptosis in the AGS cancer cell line, the synthesized MAC-NCs exhibit promising potential as a selective anti-gastric cancer agent. However, further in-vivo studies are necessary to confirm and quantify the nanoparticle's selective toxicity and pharmaceutical properties in future investigations.


Subject(s)
Alginates , Apoptosis , Cellulose , Flavonoids , Nanoparticles , Stomach Neoplasms , Humans , Alginates/chemistry , Alginates/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Apoptosis/drug effects , Nanoparticles/chemistry , Cell Line, Tumor , Cellulose/pharmacology , Cellulose/chemistry , Flavonoids/pharmacology , Caspase 9/metabolism , Caspase 9/genetics , Caspase 8/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Cell Survival/drug effects
19.
Cell Death Dis ; 15(9): 642, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227585

ABSTRACT

Paired immunoglobin-like type 2 receptor beta (PILRB) mainly plays a crucial role in regulating innate immunity, but whether PILRB is involved in cancer is poorly understood. Here, we report that PILRB potentiates the PI3K/AKT pathway to drive gastric tumorigenesis by binding and stabilizing IRS4, which could hyperactivate the PI3K/AKT pathway. Firstly, the levels of PILRB are upregulated in human gastric cancer (GC) specimens and associated with poor prognosis in patients with GC. In addition, our data show that PILRB promotes cell proliferation, colony formation, cell migration and invasion in GC cells in vitro and in vivo. Mechanistically, PILRB recruits the deubiquitination enzymes OTUB1 to IRS4 and relieves K48-linked ubiquitination of IRS4, protecting IRS4 protein from proteasomal-mediated degradation and subsequent activation of the PI3K/AKT pathway. Importantly, the levels of PILRB are positively correlated with IRS4 in GC specimens. Meanwhile, we also found that PILRB reprogrammed cholesterol metabolism by altering ABCA1 and SCARB1 expression levels, and PILRB-expression confers GC cell resistance to statin treatment. Taken together, our findings illustrate that the oncogenic role of PILRB in gastric tumorigenesis, providing new insights into the regulation of PI3K/AKT signaling in GC and establishing PILRB as a biomarker for simvastatin therapy resistance in GC.


Subject(s)
Carcinogenesis , Cholesterol , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cholesterol/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Mice , Mice, Nude , Cell Proliferation , Neoplasm Metastasis , Cell Movement , Male , Mice, Inbred BALB C
20.
BMC Cancer ; 24(1): 1130, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261761

ABSTRACT

Gastric cancer is one of the most malignant digestive tract tumors worldwide and its progression is associated with gene expression and metabolic alteration. We revealed that the gastric cancer patients with lower expression level of TOB1 exhibited poorer overall survivals according to the data in Kaplan-Meier Plotter. The unphosphorylated TOB1 protein which is effective expressed lower in gastric cancer cells. The gastric cancer cells with TOB1 gene depletion performed higher abilities of proliferation, migration and invasion and lower ability of apoptosis in vitro. The TOB1 gene depletion also promoted the tumorigenesis of gastric cancer cells in vivo. The gastric cancer cells with TOB1 gene overexpression had the converse behaviors. The transcriptional and metabolic sequencing was performed. The analyzation results showed that genes correlate-expressed with TOB1 gene were enriched in the pathways related to ERK pathway, including focal adhesion pathway, which was verified using real-time quantitative PCR. After inhibiting ERK pathway, the proliferation, colony formation and migration abilities were reduced in gastric cancer cells with low phosphorylated TOB1 protein expression level. Moreover, Pearson correlation analysis was adopted to further analyze the correlation of enriched metabolic products and differentially expressed genes. The expression of Choline, UDP-N-acetylglucosamine, Adenosine and GMP were related to the function of TOB1. This study demonstrates the genes and metabolites related to focal adhesion pathway and ERK pathway are the potential diagnosis and therapeutic targets to gastric cancer with TOB1 depletion.


Subject(s)
Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Stomach Neoplasms , Tumor Suppressor Proteins , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Cell Movement/genetics , Cell Line, Tumor , Animals , Mice , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Focal Adhesions/genetics , Focal Adhesions/metabolism , Apoptosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL