Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.279
Filter
Add more filters

Publication year range
1.
Mol Cell ; 82(4): 770-784.e9, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35114100

ABSTRACT

The mTOR complex 1 (mTORC1) is an essential metabolic hub that coordinates cellular metabolism with the availability of nutrients, including amino acids. Sestrin2 has been identified as a cytosolic leucine sensor that transmits leucine status signals to mTORC1. In this study, we identify an E3 ubiquitin ligase RING finger protein 167 (RNF167) and a deubiquitinase STAMBPL1 that function in concert to control the polyubiquitination level of Sestrin2 in response to leucine availability. Ubiquitination of Sestrin2 promotes its interaction with GATOR2 and inhibits mTORC1 signaling. Bioinformatic analysis reveals decreased RNF167 expression and increased STAMBPL1 expression in gastric and colorectal tumors. Knockout of STAMBPL1 or correction of the heterozygous STAMBPL1 mutation in a human colon cancer cell line suppresses xenograft tumor growth. Lastly, a cell-permeable peptide that blocks the STAMBPL1-Sestrin2 interaction inhibits mTORC1 and provides a potential option for cancer therapy.


Subject(s)
Colorectal Neoplasms/enzymology , Peptide Hydrolases/metabolism , Stomach Neoplasms/enzymology , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Caco-2 Cells , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred BALB C , Mice, Nude , Mutation , Nuclear Proteins/metabolism , Peptide Hydrolases/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Burden , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
Gastroenterology ; 161(6): 1907-1923.e26, 2021 12.
Article in English | MEDLINE | ID: mdl-34391772

ABSTRACT

BACKGROUND & AIMS: Metaplasia and dysplasia in the corpus are reportedly derived from de-differentiation of chief cells. However, the cellular origin of metaplasia and cancer remained uncertain. Therefore, we investigated whether pepsinogen C (PGC) transcript-expressing cells represent the cellular origin of metaplasia and cancer using a novel Pgc-specific CreERT2 recombinase mouse model. METHODS: We generated a Pgc-mCherry-IRES-CreERT2 (Pgc-CreERT2) knock-in mouse model. Pgc-CreERT2/+ and Rosa-EYFP mice were crossed to generate Pgc-CreERT2/Rosa-EYFP (Pgc-CreERT2/YFP) mice. Gastric tissues were collected, followed by lineage-tracing experiments and histologic and immunofluorescence staining. We further established Pgc-CreERT2;KrasG12D/+ mice and investigated whether PGC transcript-expressing cells are responsible for the precancerous state in gastric glands. To investigate cancer development from PGC transcript-expressing cells with activated Kras, inactivated Apc, and Trp53 signaling pathways, we crossed Pgc-CreERT2/+ mice with conditional KrasG12D, Apcflox, Trp53flox mice. RESULTS: Expectedly, mCherry mainly labeled chief cells in the Pgc-CreERT2 mice. However, mCherry was also detected throughout the neck cell and isthmal stem/progenitor regions, albeit at lower levels. In the Pgc-CreERT2;KrasG12D/+ mice, PGC transcript-expressing cells with KrasG12D/+ mutation presented pseudopyloric metaplasia. The early induction of proliferation at the isthmus may reflect the ability of isthmal progenitors to react rapidly to Pgc-driven KrasG12D/+ oncogenic mutation. Furthermore, Pgc-CreERT2;KrasG12D/+;Apcflox/flox mice presented intramucosal dysplasia/carcinoma and Pgc-CreERT2;KrasG12D/+;Apcflox/flox;Trp53flox/flox mice presented invasive and metastatic gastric carcinoma. CONCLUSIONS: The Pgc-CreERT2 knock-in mouse is an invaluable tool to study the effects of successive oncogenic activation in the mouse corpus. Time-course observations can be made regarding the responses of isthmal and chief cells to oncogenic insults. We can observe stomach-specific tumorigenesis from the beginning to metastatic development.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/genetics , Chief Cells, Gastric/enzymology , Integrases/genetics , Pepsinogen C/genetics , Precancerous Conditions/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Stomach Neoplasms/genetics , Transcriptional Activation , Animals , Cell Dedifferentiation , Cell Lineage , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chief Cells, Gastric/pathology , Gene Expression Regulation, Neoplastic , Genes, APC , Genetic Predisposition to Disease , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Metaplasia , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Pepsinogen C/metabolism , Phenotype , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Red Fluorescent Protein
3.
J Clin Lab Anal ; 36(5): e24313, 2022 May.
Article in English | MEDLINE | ID: mdl-35313389

ABSTRACT

BACKGROUND: Kinesin family member 2A (KIF2A) induces gastric cancer (GC) growth and invasion, while its clinical relevance in GC patients is not reported. This study aimed to investigate the linkage of KIF2A with clinicopathological features, prognosis, and chemosensitivity of GC. METHODS: A total of 160 surgical GC patients were reviewed, with their tumor and adjacent tissues acquired for immunohistochemical (IHC) assay to measure KIF2A expression, then scored by a semi-quantitative method (IHC score: 0-12). KIF2A siRNA or nonsense-siRNA were transfected into HGC-27 and NCI-N87 cells underwent various concentrations of capecitabine or oxaliplatin treatment followed by chemosensitivity assessment. RESULTS: Kinesin family member 2A expression was elevated in the tumor tissue compared to the adjacent tissue (IHC score: 5.6 ± 3.1 vs. 2.9 ± 1.7, p < 0.001). Besides, tumor KIF2A expression was related to larger tumor size (p = 0.014), higher N stage (p = 0.004) and TNM stage (p = 0.011); however, it was not linked with other clinicopathological features (all p > 0.05). Signally, tumor KIF2A high expression predicted poor overall survival (p = 0.037). After adjustment via multivariate Cox's regression, tumor KIF2A high expression independently linked with worse disease-free survival (p = 0.033). Finally, KIF2A knockdown improved the oxaliplatin chemosensitivity vastly but only slightly affected capecitabine chemosensitivity in HGC-27 and NCI-N87 cells. CONCLUSION: Kinesin family member 2A reflects larger tumor size, advanced TNM stage, improved chemosensitivity, and predicts unfavorable survival in GC.


Subject(s)
Kinesins , Stomach Neoplasms , Capecitabine/pharmacology , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Kinesins/genetics , Kinesins/metabolism , Oxaliplatin/pharmacology , Prognosis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics
4.
J Cell Physiol ; 236(4): 2740-2755, 2021 04.
Article in English | MEDLINE | ID: mdl-32914432

ABSTRACT

Gastric cancer (GC) is one of the most common malignant tumors in China and the third leading cause of cancer-related death. Parkin has been shown to be a tumor suppressor in a variety of malignancies, including GC. However, the mechanism of Parkin in GC remains unclear. In this study, the low expression of Parkin in GC cells and patient tumor tissues was observed, and Parkin inhibited proliferation and migration of GC cells. Additionally, doxorubicin (DOX) upregulated the expression of Parkin and promoted its anticancer effect. Forkhead box O3 (FOXO3a) is a crucial transcription factor that involves in the regulation of cancer cell proliferation, apoptosis, and metabolism. Here, we found that FOXO3a inhibits cell proliferation, migration, and promotes apoptosis in GC by regulating Parkin expression at the transcriptional level. In addition, Parkin inhibited cell proliferation, migration, and promoted apoptosis by inhibiting ATP-binding box protein E1 (ABCE1) expression. In summary, our results demonstrated a new regulatory axis of FOXO3a-Parkin-ABCE1 that modulated GC cell proliferation, migration, and apoptosis, and it can serve as a potential therapeutic target in GC.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Forkhead Box Protein O3/metabolism , Stomach Neoplasms/enzymology , Ubiquitin-Protein Ligases/metabolism , ATP-Binding Cassette Transporters/genetics , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Doxorubicin/pharmacology , Forkhead Box Protein O3/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics
5.
J Cell Biochem ; 122(10): 1376-1388, 2021 10.
Article in English | MEDLINE | ID: mdl-34160883

ABSTRACT

Gastric cancer is one of the most common and deadly types of cancer in the world, and poor prognosis with treatment failure is widely reported in the literature. In this context, kinases have been considered a relevant choice for targeted therapy in gastric cancer. Here, we explore the antiproliferative and antimigratory effects of the AURKA inhibitor and the prognostic and therapeutic value as a biomarker of gastric cancer. A total of 145 kinase inhibitors were screened to evaluate the cytotoxic or cytostatic effects in the gastric cancer cell line. Using the Alamar Blue assay, flow cytometry, quantitative polymerase chain reaction, and observation of caspase 3/7 activity and cell migration, we investigated the antiproliferative, proapoptotic, and antimigratory effects of the AURKA inhibitor. Moreover, AURKA overexpression was evaluated in the gastric cell lines and the gastric tumor tissue. Out of the 145 inhibitors, two presented the highest antiproliferative effect. Both molecules can induce apoptosis by the caspases 3/7 pathway in addition to inhibiting cancer cell migration, mainly the AURKA inhibitor. Moreover, molecular docking analysis revealed that GW779439X interacts in the active site of the AURKA enzyme with similar energy as a well-described inhibitor. Our study identified AURKA overexpression in the gastric cancer cell line and gastric tumor tissue, revealing that its overexpression in patients with cancer is correlated with low survival. Therefore, it is feasible to suggest AURKA as a potential marker of gastric cancer, besides providing robust information for diagnosis and estimated survival of patients. AURKA can be considered a new molecular target used in the prognosis and therapy of gastric cancer.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridazines/pharmacology , Stomach Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Apoptosis , Aurora Kinase A/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , High-Throughput Screening Assays , Humans , Male , Middle Aged , Molecular Docking Simulation , Prognosis , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Survival Rate
6.
Cancer Sci ; 112(5): 1936-1942, 2021 May.
Article in English | MEDLINE | ID: mdl-33453146

ABSTRACT

Extrapulmonary neuroendocrine carcinoma (EPNEC) is a lethal disease with a poor prognosis. Platinum-based chemotherapy is used as the standard first-line treatment for unresectable EPNEC. Several retrospective studies have reported the results of the utilization of temozolomide (TMZ) as a drug for the second-line treatment for EPNEC. Patients with unresectable EPNEC that were resistant to platinum-based combination chemotherapy were recruited for a prospective phase II study of TMZ monotherapy. A 200 mg/m2 dose of TMZ was given from day 1 to day 5, every 4 weeks. Response rate (RR) was evaluated as the primary end-point. The presence of O6 -methylguanine DNA methyltransferase (MGMT) in EPNEC patients was also evaluated as exploratory research. Thirteen patients were enrolled in this study. Primary lesions were pancreas (n = 3), stomach (n = 3), duodenum (n = 1), colon (n = 1), gallbladder (n = 1), liver (n = 1), uterus (n = 1), bladder (n = 1), and primary unknown (n = 1). Each case was defined as pathological poorly differentiated neuroendocrine carcinoma from surgically resected and/or biopsied specimens. The median Ki-67 labeling index was 60% (range, 22%-90%). The RR was 15.4%, progression-free survival was 1.8 months (95% confidence interval [CI], 1.0-2.7), overall survival (OS) was 7.8 months (95% CI, 6.0-9.5), and OS from first-line treatment was 19.2 months (95% CI, 15.1-23.3). No grade 3 or 4 hematological toxicity had occurred and there was one case of grade 3 nausea. One case presented MGMT deficiency and this case showed partial response. Temozolomide monotherapy is a feasible, modestly effective, and safe treatment for patients with unresectable EPNEC following platinum-based chemotherapy, especially those with MGMT deficiency.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Temozolomide/therapeutic use , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Drug Administration Schedule , Duodenal Neoplasms/drug therapy , Duodenal Neoplasms/enzymology , Duodenal Neoplasms/pathology , Female , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/enzymology , Gallbladder Neoplasms/pathology , Humans , Ki-67 Antigen/analysis , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/analysis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Progression-Free Survival , Prospective Studies , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Temozolomide/administration & dosage , Temozolomide/adverse effects , Uterine Neoplasms/drug therapy , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology
7.
Biochem Biophys Res Commun ; 540: 108-115, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33476960

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has the ability to selectively trigger cancer cell apoptosis and can be used as a target for tumor therapy. However, gastric cancer cells are usually insensitive to TRAIL so reducing this drug resistance may improve the treatment of gastric cancer. In this study, we used Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) experiments to determine the effects of 5-fluorouracil (5-FU) and TRAIL on the proliferation of gastric cancer cells. An Annexin V/propidium iodide (PI) staining experiment was used to detect apoptosis, and Western blotting was used to analyze the expression levels of apoptosis-related proteins and mitogen-activated protein kinase (MAPK) pathway proteins. The antitumor effects of 5-FU and TRAIL were verified in vivo using a nude mouse tumorigenesis experiment, and a TUNEL assay was performed to evaluate apoptosis in tumor tissue from the nude mice. We found the combination of 5-FU and TRAIL had a greater inhibitory effect on the proliferation of gastric cancer cells than 5-FU or TRAIL alone both in vivo and in vitro. 5-FU enhanced TRAIL-induced gastric cancer cell apoptosis by inactivating the MAPK pathway. Overall, our analysis firstly provided new insights into the role of 5-FU in increasing sensitivity to TRAIL. 5-FU can be used as a sensitizer for TRAIL, and its administration is a potential strategy for the treatment of gastric cancer.


Subject(s)
Fluorouracil/pharmacology , Fluorouracil/therapeutic use , MAP Kinase Signaling System/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Nude , Stomach Neoplasms/enzymology
8.
Mod Pathol ; 34(3): 672-683, 2021 03.
Article in English | MEDLINE | ID: mdl-32973329

ABSTRACT

Although a certain proportion of intramucosal carcinomas (IMCs) of the stomach does metastasize, the majority of patients are currently treated with endoscopic resection without lymph node dissection, and this potentially veils any existing metastasis and may put some patients in danger. In this regard, biological markers from the resected IMC that can predict metastasis are warranted. Here, we discovered unique miRNA expression profiles that consist of 21 distinct miRNAs that are specifically upregulated (miR-628-5p, miR-1587, miR-3175, miR-3620-5p, miR-4459, miR-4505, miR-4507, miR-4720-5p, miR-4742-5p, and miR-6779-5p) or downregulated (miR-106b-3p, miR-125a-5p, miR-151b, miR-181d-5p, miR-486-5p, miR-500a-3p, miR-502-3p, miR-1231, miR-3609, and miR-6831-5p) in metastatic (M)-IMC compared to nonmetastatic (N)-IMC, or nonneoplastic gastric mucosa. Intriguingly, most of these selected miRNAs showed stepwise increased or decreased expression from nonneoplastic tissue to N-IMC to M-IMC. This suggests that common oncogenic mechanisms are gradually intensified during the metastatic process. Using a machine-learning algorithm, we demonstrated that such miRNA signatures could distinguish M-IMC from N-IMC. Gene ontology and pathway analysis revealed that TGF-ß signaling was enriched from upregulated miRNAs, whereas E2F targets, apoptosis-related, hypoxia-related, and PI3K/AKT/mTOR signaling pathways, were enriched from downregulated miRNAs. Immunohistochemical staining of samples from multiple institutions indicated that PI3K/AKT/mTOR pathway components, MAPK1, phospho-p44/42 MAPK, and pS6 were highly expressed and the expression of SMAD7, a TGF-ß pathway component, was decreased in M-IMC, which could aid in distinguishing M-IMC from N-IMC. The miRNA signature discovered in this study is a valuable biological marker for identifying metastatic potential of IMCs, and provides novel insights regarding the metastatic progression of IMC.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Transcriptome , Biomarkers, Tumor/analysis , Gastric Mucosa/enzymology , Gastric Mucosa/pathology , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunohistochemistry , Lymphatic Metastasis , Machine Learning , Predictive Value of Tests , Reproducibility of Results , Republic of Korea , Signal Transduction/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
9.
Pharmacol Res ; 170: 105493, 2021 08.
Article in English | MEDLINE | ID: mdl-33600940

ABSTRACT

Gastric carcinoma has serious morbidity and mortality, which seriously threats human health. The studies on gastrointestinal cell biology have shown that the ubiquitination modification that occurs after protein translation plays an essential role in the pathogenesis of gastric carcinoma. Protein ubiquitination is catalyzed by E3 ubiquitin ligase and can regulate various substrate proteins in different cellular pathways. Cullin-RING E3 ligase (CRLs) is a representative of the E3 ubiquitin ligase family, which requires cullin (CUL) neddylation modification for activation to regulate homeostasis of ~20% of cellular proteins. The substrate molecules regulated by CRLs are often involved in many cell progressions such as cell cycle progression, cell apoptosis, DNA damage and repair. Given that CRLs play an important role in modulation of biological activities, so targeting a certain CULs member neddylation may be an attractive strategy for selectively controlling the cellular proteins levels to achieve the goal of cancer treatment. In this review, we will discuss the roles of CULs and Ring protein in gastric carcinoma and summarize the current neddylation modulators for gastric carcinoma treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Cullin Proteins/metabolism , Enzyme Inhibitors/therapeutic use , Stomach Neoplasms/drug therapy , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitins/metabolism , Animals , Antineoplastic Agents/adverse effects , Carcinoma/enzymology , Carcinoma/pathology , Enzyme Inhibitors/adverse effects , Humans , Molecular Targeted Therapy , Proteolysis , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
10.
Dig Dis Sci ; 66(12): 4290-4301, 2021 12.
Article in English | MEDLINE | ID: mdl-33449226

ABSTRACT

BACKGROUND: Circular RNA (circRNA) has been shown to be closely associated with cancer progression, including gastric cancer (GC). However, the function of circ_0004104 in GC progression has not been clarified. AIMS: The purpose of this study was to explore the role of circ_0004104 in GC progression. METHODS: The expression levels of circ_0004104, miR-539-3p, and ring finger protein 2 (RNF2) were assessed using quantitative real-time polymerase chain reaction. Cell proliferation was measured by 3-(4,5-dimethyl-2 thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, and cell migration and invasion were detected using transwell assay. The levels of glutamine, glutamate, and α-ketoglutarate were determined to evaluate the glutaminolysis of cells, and the protein levels of glutaminolysis-related markers and RNF2 were detected using western blot analysis. Furthermore, Dual-Luciferase Reporter Assay was employed to assess the interaction between miR-539-3p and circ_0004104 or RNF2. Animal experiments were carried out to evaluate the effect of circ_0004104 silencing on GC tumor growth in vivo. RESULTS: Circ_0004104 was upregulated in GC, and its knockdown repressed the proliferation, metastasis, and glutaminolysis of GC cells in vitro and reduced GC tumor growth in vivo. Furthermore, we discovered that circ_0004104 could sponge miR-539-3p and miR-539-3p could target RNF2. The rescue experiments suggested that miR-539-3p inhibitor could reverse the suppressive effect of circ_0004104 silencing on GC progression, and RNF2 overexpression also reversed the inhibition effect of miR-539-3p mimic on GC progression. CONCLUSION: Circ_0004104 accelerated GC progression via regulating the miR-539-3p/RNF2 axis, indicating that circ_0004104 might be a potential therapeutic target for GC.


Subject(s)
Exosomes/metabolism , MicroRNAs/metabolism , Polycomb Repressive Complex 1/metabolism , RNA, Circular/metabolism , Stomach Neoplasms/enzymology , Aged , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Exosomes/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Polycomb Repressive Complex 1/genetics , RNA, Circular/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Burden
11.
Dig Dis Sci ; 66(12): 4274-4289, 2021 12.
Article in English | MEDLINE | ID: mdl-33449227

ABSTRACT

BACKGROUND: Exosome-mediated transfer of circular RNAs (circRNAs) is related to gastric cancer (GC) development. CircRNA NIMA-related kinase 9 (circNEK9; hsa_circ_0032683) was reported to be up-regulated in GC. AIMS: The biological role of circNEK9 and its underlying mechanisms in GC progression were explored. METHODS: The levels of RNAs and proteins were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assay. Cell proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and flow cytometry. Wound healing assay and transwell assays were conducted to analyze cell motility. Intermolecular interaction was verified by dual-luciferase reporter assay and RNA pull-down assay. Animal experiments were used to evaluate the role of circNEK9 in the growth of xenograft tumors in vivo. RESULTS: CircNEK9 was up-regulated in GC tissues and cell lines. CircNEK9 interference suppressed the proliferation and motility of GC cells. CircNEK9 silencing enhanced microRNA-409-3p (miR-409-3p) level through direct interaction. CircNEK9 silencing-mediated influences on the proliferation and metastasis of GC cells were partly overturned by the interference of miR-409-3p. MiR-409-3p directly interacted with microtubule-associated protein 7 (MAP7) messenger RNA (mRNA). MiR-409-3p-induced effects in GC cells were largely counteracted by the overexpression of MAP7. CircNEK9 silencing blocked GC tumor growth in vivo. Exosome-mediated transfer of circNEK9 promoted the motility of recipient GC cells. CONCLUSIONS: CircNEK9 accelerated the proliferation, migration, and invasion of GC cells through targeting miR-409-3p/MAP7 axis. Plasma exosomal circNEK9 promoted the migration and invasion of recipient GC cells.


Subject(s)
Exosomes/metabolism , MicroRNAs/metabolism , Microtubule-Associated Proteins/metabolism , RNA, Circular/metabolism , Stomach Neoplasms/enzymology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Exosomes/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasm Invasiveness , RNA, Circular/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Burden
12.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1216-1226, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34363072

ABSTRACT

Glycosylation is a common posttranslational modification of proteins, which plays a role in the malignant transformation, growth, progression, chemoresistance, and immune response of tumors. Disulfide isomerase family A3 (PDIA3) specifically acts on newly synthesized glycoproteins to promote the correct folding of sugar chains. Studies have shown that PDIA3 participates in multidrug-resistant gastric cancer (MDR-GC). In this study, we performed western blot analysis and immunohistochemistry to identify PDIA3 expression. Cell proliferation was assessed by CCK-8 assay. Transwell assays were used to detect the migration and invasion abilities of cells. Immunoprecipitation coupled to mass spectrometry (IP-MS) analysis was employed to identify PDIA3-interacting proteins and the associated pathways in MDR-GC cells. Glycoprotein interactions and translocation were detected by immunofluorescence assay. The results showed that PDIA3 knockdown significantly inhibited the proliferation, invasion, and migration abilities of MDR-GC cells. Kyoto Encyclopedia of Genes and Genomes analysis of the IP-MS results showed that PDIA3 was closely associated with focal adhesion pathways in MDR-GC cells. Additionally, important components of focal adhesion pathways, including fibronectin-1 (FN1) and integrin α5 (ITGA5), were identified as pivotal PDIA3-binding glycoproteins. Knockdown of PDIA3 altered the cellular locations of FN1 and ITGA5, leading to abnormal accumulation. In conclusion, our results suggest that knockdown of PDIA3 inhibited the malignant behaviors of MDR-GC cells and influenced the translocation of FN1 and ITGA5.


Subject(s)
Cell Proliferation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Endoplasmic Reticulum/enzymology , Gene Silencing , Neoplasm Proteins/biosynthesis , Protein Disulfide-Isomerases/biosynthesis , Stomach Neoplasms/enzymology , Cell Line, Tumor , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Humans , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Protein Disulfide-Isomerases/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
13.
Biochem Genet ; 59(3): 652-667, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33442814

ABSTRACT

As the endogenous ligand for the GH secretagogue receptor (GHSR), Ghrelin is aberrant expressed in multiple malignant carcinoma, and involved in regulating a number of progression of cancer, especially in metastasis and proliferation. However, the precise role of Ghrelin in tumorigenesis of gastric cancer (GC) is still poorly understood. In this study, we extensively investigated the roles and mechanisms of Ghrelin in human gastric cancer. Ghrelin levels in cancer tissues and cell lines were analyzed by immunohistochemistry, qRT-PCR, and Western blot. Functional studies were performed after Ghrelin overexpressed or knockdown in AGS cell line. Cell proliferation was evaluated in by MTT and clone formation assays. The wound healing and Transwell system were used to assess the cell migration and invasive ability of GC cells. Cell apoptosis was detected by flow cytometry, and metabolic assays were performed to reveal the function of Warburg effect in the process. Ghrelin was lowly expressed in gastric cancer tissues and cell lines. Overexpression of Ghrelin inhibited gastric cancer cell proliferation, migration, invasion, and promoted apoptosis by activating the AMPK pathway, while D-[lys3]-GHRP-6 (a GHSR agonist) treatment relieved the effect, promoting tumorigenesis. Ghrelin knockdown increased the glucose uptake and lactic acid release, suggesting that Ghrelin elicited an anti-Warburg effect via AMPK pathway to inhibit gastric tumorigenesis. Ghrelin inhibits cell proliferation, migration, and invasion by eliciting an anti-Warburg effect via AMPK signaling pathway in gastric cancer cells.


Subject(s)
Adenylate Kinase/metabolism , Ghrelin/physiology , Signal Transduction , Stomach Neoplasms/pathology , Apoptosis/physiology , Carcinogenesis , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/physiology , Disease Progression , Down-Regulation , Ghrelin/antagonists & inhibitors , Ghrelin/metabolism , Glucose/metabolism , Humans , Neoplasm Metastasis , Stomach Neoplasms/enzymology , Stomach Neoplasms/metabolism , Up-Regulation
14.
Drug Dev Res ; 82(8): 1182-1192, 2021 12.
Article in English | MEDLINE | ID: mdl-34002415

ABSTRACT

The oncogenic signaling pathway Wnt is often activated in many cancers including gastric cancer. Wnt signaling pathway is considered as a potential target for developing new targeted therapeutics. Kinase inhibitors are the promising class of drugs for many diseases including cancers. Toward identifying the potent inhibitors targeting Wnt signaling pathway, a kinase inhibitor library with 82 inhibitors were screened using Wnt pathway reporter assay in gastric cancer cells. Notably, 34 kinase inhibitors were identified to inhibit Wnt mediated reporter activity to the extent of more than 50%. The corresponding kinase genes, which are known targets of these kinase inhibitors, were investigated for their expression in the available mRNA profiles of gastric tumors. A major group of the kinase genes showed higher expression in intestinal subtype gastric tumors. Another group of kinase genes were found expressed in diffuse type gastric tumors. The kinase genes expressed in intestinal type gastric tumors were found associated with varying survival of gastric cancer patients whereas those expressed in diffuse type tumors were found associated with the poor survival. Thus, the kinase genes specifically expressed in intestinal and diffuse type gastric tumors and the kinase inhibitors to target Wnt signaling pathway in gastric cancer subtypes have been identified.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Stomach Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Cell Line, Tumor , Humans , Protein Kinase Inhibitors/therapeutic use , Stomach Neoplasms/enzymology , Stomach Neoplasms/mortality
15.
Molecules ; 26(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809289

ABSTRACT

Matrix metalloproteinases (MMPs), key molecules of cancer invasion and metastasis, degrade the extracellular matrix and cell-cell adhesion molecules. MMP-10 plays a crucial role in Helicobacter pylori-induced cell-invasion. The mitogen-activated protein kinase (MAPK) signaling pathway, which activates activator protein-1 (AP-1), is known to mediate MMP expression. Infection with H. pylori, a Gram-negative bacterium, is associated with gastric cancer development. A toxic factor induced by H. pylori infection is reactive oxygen species (ROS), which activate MAPK signaling in gastric epithelial cells. Peroxisome proliferator-activated receptor γ (PPAR-γ) mediates the expression of antioxidant enzymes including catalase. ß-Carotene, a red-orange pigment, exerts antioxidant and anti-inflammatory properties. We aimed to investigate whether ß-carotene inhibits H. pylori-induced MMP expression and cell invasion in gastric epithelial AGS (gastric adenocarcinoma) cells. We found that H. pylori induced MMP-10 expression and increased cell invasion via the activation of MAPKs and AP-1 in gastric epithelial cells. Specific inhibitors of MAPKs suppressed H. pylori-induced MMP-10 expression, suggesting that H. pylori induces MMP-10 expression through MAPKs. ß-Carotene inhibited the H. pylori-induced activation of MAPKs and AP-1, expression of MMP-10, and cell invasion. Additionally, it promoted the expression of PPAR-γ and catalase, which reduced ROS levels in H. pylori-infected cells. In conclusion, ß-carotene exerts an inhibitory effect on MAPK-mediated MMP-10 expression and cell invasion by increasing PPAR-γ-mediated catalase expression and reducing ROS levels in H. pylori-infected gastric epithelial cells.


Subject(s)
Gastric Mucosa/drug effects , Helicobacter pylori/pathogenicity , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , beta Carotene/pharmacology , Catalase/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/microbiology , Gastric Mucosa/enzymology , Gastric Mucosa/microbiology , Gene Expression Regulation, Enzymologic/drug effects , Helicobacter Infections/complications , Helicobacter Infections/enzymology , Helicobacter Infections/pathology , Helicobacter pylori/drug effects , Humans , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 10/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , PPAR gamma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Stomach Neoplasms/enzymology , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Transcription Factor AP-1/metabolism
16.
J Cell Mol Med ; 24(19): 11133-11145, 2020 10.
Article in English | MEDLINE | ID: mdl-32827244

ABSTRACT

Acylglycerol kinase (AGK) uses adenosine triphosphate (ATP) and acylglycerol to generate adenosine diphosphate (ADP) and acyl-sn-glycerol 3-phosphate in cells. Recent evidence has demonstrated that dysregulated AGK expression is associated with the development of various human cancers. This study investigated the effects of AGK on gastric cancer cell proliferation and carcinogenesis and explored the underlying molecular events. AGK expression was up-regulated in gastric cancer and was associated with poor prognosis in gastric cancer patients. AGK overexpression increased gastric cancer proliferation, invasion capacity and the expression of the epithelial-mesenchymal transition markers in vitro. Conversely, the knockdown of AGK expression reduced gastric cancer cell proliferation in vitro and in nude mouse tumour cell xenografts. Importantly, AGK expression was associated with the YAP1 expression in gastric cancer cells and tissues. YAP1 expression also transcriptionally induced AGK expression through the binding of TEAD to the AGK gene promoter. However, AGK expression inhibited the activation of the Hippo pathway proteins and induced YAP1 nuclear localization to enhance the transcription activity of YAP1/TEADs. In conclusion, the study demonstrates that AGK is not only a novel target of the Hippo-YAP1 pathway, but that it also positively regulates YAP1 expression, thus forming a YAP1-AGK-positive feedback loop.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Phosphotransferases (Alcohol Group Acceptor)/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Transcription Factors/metabolism , Up-Regulation/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Mice, Nude , Neoplasm Invasiveness , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Stomach Neoplasms/pathology , Transcription, Genetic , YAP-Signaling Proteins
17.
J Biol Chem ; 294(20): 8238-8258, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30940726

ABSTRACT

The subcellular mechanism by which nonsteroidal anti-inflammatory drugs (NSAIDs) induce apoptosis in gastric cancer and normal mucosal cells is elusive because of the diverse cyclooxygenase-independent effects of these drugs. Using human gastric carcinoma cells (AGSs) and a rat gastric injury model, here we report that the NSAID indomethacin activates the protein kinase Cζ (PKCζ)-p38 MAPK (p38)-dynamin-related protein 1 (DRP1) pathway and thereby disrupts the physiological balance of mitochondrial dynamics by promoting mitochondrial hyper-fission and dysfunction leading to apoptosis. Notably, DRP1 knockdown or SB203580-induced p38 inhibition reduced indomethacin-induced damage to AGSs. Indomethacin impaired mitochondrial dynamics by promoting fissogenic activation and mitochondrial recruitment of DRP1 and down-regulating fusogenic optic atrophy 1 (OPA1) and mitofusins in rat gastric mucosa. Consistent with OPA1 maintaining cristae architecture, its down-regulation resulted in EM-detectable cristae deformity. Deregulated mitochondrial dynamics resulting in defective mitochondria were evident from enhanced Parkin expression and mitochondrial proteome ubiquitination. Indomethacin ultimately induced mitochondrial metabolic and bioenergetic crises in the rat stomach, indicated by compromised fatty acid oxidation, reduced complex I- associated electron transport chain activity, and ATP depletion. Interestingly, Mdivi-1, a fission-preventing mito-protective drug, reversed indomethacin-induced DRP1 phosphorylation on Ser-616, mitochondrial proteome ubiquitination, and mitochondrial metabolic crisis. Mdivi-1 also prevented indomethacin-induced mitochondrial macromolecular damage, caspase activation, mucosal inflammation, and gastric mucosal injury. Our results identify mitochondrial hyper-fission as a critical and common subcellular event triggered by indomethacin that promotes apoptosis in both gastric cancer and normal mucosal cells, thereby contributing to mucosal injury.


Subject(s)
Apoptosis/drug effects , GTP Phosphohydrolases/metabolism , Gastric Mucosa/enzymology , Indomethacin/pharmacology , MAP Kinase Signaling System/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondria/enzymology , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Kinase C/metabolism , Stomach Neoplasms/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Dynamins , GTP Phosphohydrolases/genetics , Gastric Mucosa/pathology , Humans , MAP Kinase Signaling System/genetics , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Neoplasm Proteins/genetics , Protein Kinase C/genetics , Rats , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/genetics
18.
J Cell Physiol ; 235(5): 4709-4719, 2020 05.
Article in English | MEDLINE | ID: mdl-31650535

ABSTRACT

Studies have found that miR-665 acted as a tumor suppressor or an oncogene in different malignancies. miR-665 expression was elevated in gastric adenocarcinoma tissues; however, its role and mechanism in this disease are not fully clarified. The expression of miR-665 and its target gene was detected in human gastric adenocarcinoma tissues and cells. Moreover, we analyzed the effects of miR-665 on the proliferation, migration, and epithelial-mesenchymal transition (EMT) of gastric adenocarcinoma cells as well as tumor growth in vivo. The mechanisms of miR-665 in gastric adenocarcinoma were investigated by using molecular biology techniques. We found miR-665 was upregulated and suppressor of cytokine signaling 3 (SOCS3) was downregulated in gastric adenocarcinoma tissues and cells. Elevated miR-665 was positively correlated with tumor size, lymph node metastasis, invasion depth, TNM stage, and poor differentiation in gastric adenocarcinoma patients. Overexpression of miR-665 promoted, whereas knockdown of miR-665 suppressed the proliferation, migration, and EMT of gastric adenocarcinoma cells. Furthermore, we demonstrated that miR-665 functioned through targeting SOCS3, followed by activation of the FAK/Src signaling pathway in gastric adenocarcinoma cells. miR-665 antagomir inhibited tumor growth as well as the activation of the FAK/Src pathway but increased SOCS3 expression in nude mice. In addition, miR-665 expression was negatively regulated by long noncoding RNA maternally expressed gene 3 (MEG3). In conclusion, miR-665 acted as an oncogene in gastric adenocarcinoma by inhibiting SOCS3 followed by activation of the FAK/Src pathway and it was negatively modulated by MEG3. miR-665 may be a promising therapeutic target for the treatment of gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/enzymology , Focal Adhesion Kinase 1/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Stomach Neoplasms/enzymology , Suppressor of Cytokine Signaling 3 Protein/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Enzyme Activation , Epithelial-Mesenchymal Transition , Female , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , MicroRNAs/genetics , Middle Aged , RNA, Long Noncoding/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Suppressor of Cytokine Signaling 3 Protein/genetics , Tumor Burden
19.
Am J Physiol Gastrointest Liver Physiol ; 318(1): G10-G22, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31433213

ABSTRACT

Gastric cancer (GC) is one of the most common cancers in the world and remains a heavy burden of health worldwide. Adenylate cyclase 3 (ADCY3) is a widely expressed membrane-associated protein in human tissues and has been identified to be a new molecular target of GC. Long noncoding RNAs have a substantial influence on tumorigenesis and progression of tumors by binding to microRNAs. Therefore, this study is to clarify the mechanism by which LINC00319 sponges micro RNA-335-5p (miR-335-5p) to influence the development of GC. Initially, microarray analysis identified GC-related differentially expressed LINC00319 and ADCY3 for this study. The interaction was confirmed that LINC00319 interacted with miR-335-5p to regulate ADCY3. Next, SGC-7901 cells presenting with the lowest LINC00319 expression and the highest miR-335-5p expression were transfected with LINC00319, miR-335-5p inhibitor, or ADCY3 vector to examine their roles in growth and metastasis of GC cells, which was further ascertained by in vivo experiments. LINC00319 was upregulated and miR-335-5p was downregulated in GC cells. LINC00319 overexpression, miR-335-5p inhibitor, or ADCY3 overexpression was shown to significantly elevate the expression of cyclin-dependent kinase 4 and metastasis associated 1, decrease that of growth arrest-specific 1, and promote tumor growth and metastasis by increasing proliferation and migration and reducing cell apoptosis. Importantly, it was found that overexpressed miR-335-5p exerted its tumor suppressive role in GC through downregulating ADCY3. Collectively, LINC00319 expedited growth and metastasis of GC by upregulating miR-335-5p-mediated ADCY3.NEW & NOTEWORTHY This study is carried out based on in vivo and in vitro studies in mice and gastric cancer (GC) cells with the aim of clarifying the role of LINC00319 on GC growth and metastasis, which associated with micro RNA-335-5p-mediated adenylate cyclase 3. Altogether, we identified LINC00319 to be a potential therapy to treat GC.


Subject(s)
Adenylyl Cyclases/metabolism , Cell Movement , Cell Proliferation , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Stomach Neoplasms/enzymology , 3' Untranslated Regions , Adenylyl Cyclases/genetics , Animals , Apoptosis , Binding Sites , Cell Line, Tumor , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Databases, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Burden , Up-Regulation
20.
Biochem Biophys Res Commun ; 521(4): 1003-1009, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31727367

ABSTRACT

Helicobacter pylori (H. pylori) infection can promote the development of gastric cancer (GC); however, the underlying mechanism is not clear. FAM60A has been found showing high levels in some cancer cells, including lung cancer (A549), and pancreatic cancer (Capan-2) cell lines. Data in oncomine showed that FAM60A overexpression was an critical prognostic factor in GC. In this study, we showed that knockdown of FAM60A could revert the increase of proliferation and the decrease of apoptosis caused by H.pylori infection in HGC-27 and AGS cells. Conversely, FAM60A upregulation promoted proliferation and inhibited apoptosis in HGC-27 and AGS cells. We also found that the PI3K/AKT pathway inhibitor LY294002 could revert the changes caused by FAM60A upregulation in HGC-27 and AGS cells. Thus, our study provides evidence that FAM60A act as a carcinogen and suggests that H. pylori-induced upregulation of FAM60A may contribute to the development of gastric cancer.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/metabolism , Helicobacter pylori/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adult , Aged , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Helicobacter Infections/genetics , Humans , Male , Middle Aged , Prognosis , Signal Transduction , Stomach Neoplasms/enzymology , Stomach Neoplasms/microbiology , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL