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










Database
Language
Publication year range
1.
Aging (Albany NY) ; 13(7): 9766-9779, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33744848

ABSTRACT

As biomolecules of great clinical value, lncRNAs play a crucial role as regulators in the processes of tumor origin, metastasis, and recurrence. Thus, lncRNAs are urgently needed for research in gastric cancer. We elucidated the specific function of OGFRP1, both in vitro and in vivo. OGFRP1 was expressed at abnormally high levels in gastric cancer samples (n = 408) compared to normal samples (n = 211). Similar results were obtained in 30 clinical case samples. Interference of OGFRP1 markedly blocked cell proliferation and migration, and it induced cell cycle arrest and the apoptosis of gastric cancer cells in vitro. Phosphorylation of AKT was inhibited in cells transfected with OGFRP1 siRNA, as compared to their control cells. The in vivo results further confirmed the antitumor effects of OGFRP1 knockdown on gastric cancer. Decreases in tumor volume (104.23±62.27 mm3) and weight (0.1006±0.0488 g) in nude mice were observed during the OGFRP1 interference, as compared with the control group (418.96±211.96 mm3 and 0.2741±0.0769 g). OGFRP1 promotes tumor progression through activating the AKT/mTOR pathway. Our findings provide a new potential target for the clinical treatment of human gastric cancer.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gastric Mucosa/metabolism , Gene Knockdown Techniques , Humans , Mice , Phosphorylation , RNA, Long Noncoding/genetics , Signal Transduction/physiology , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
2.
Onco Targets Ther ; 13: 5657-5668, 2020.
Article in English | MEDLINE | ID: mdl-32606779

ABSTRACT

PURPOSE: KIAA1522 was previously identified to play a crucial role in cancer development and progression. However, its functions and underlying mechanisms in hepatocellular carcinoma (HCC) remain elusive. MATERIALS AND METHODS: To elucidate the role of KIAA1522 in HCC, its expression was assessed using The Cancer Genome Atlas and GEPIA databases. Next, these results were validated by quantitative reverse transcription-polymerase chain reaction, Western blotting, and immunohistochemistry of HCC tissues and cell lines. Flow cytometry, CCK-8, EDU, colony formation, Transwell invasion, and wound healing assays were performed to explore the function of KIAA1522 in HCC in vivo and in vitro. Finally, gene set enrichment analysis was used to identify the pathways involved. RESULTS: Our results demonstrated that KIAA1522 was highly expressed in HCC tissues and cell lines. Furthermore, KIAA1522 overexpression was associated with unfavorable clinicopathological characteristics. Survival analyses revealed that KIAA1522 overexpression predicted lower recurrence-free and overall survival rates in patients with HCC. Functional studies suggested that KIAA1522 facilitated HCC proliferation, migration, and invasion both in vitro and in vivo. Moreover, KIAA1522 up-regulated the Wnt/ß-catenin signaling pathway, as confirmed by TOP-flash/FOP-flash luciferase reporter assays and Western blotting. CONCLUSION: In conclusion, we highlighted the oncogenic role of KIAA1522 in HCC and determined its potential as a therapeutic target for HCC.

3.
Dose Response ; 18(2): 1559325820918761, 2020.
Article in English | MEDLINE | ID: mdl-32341683

ABSTRACT

The objective of this study was to achieve the practical bioapplications of silicon nanowires (SiNWs). In this study, the tumor and normal cell lines were used as models to systematically investigate the cytotoxicity of SiNWs synthesized by HF-assisted etching methods. Morphology observation, Cell Counting Kit 8, real-time polymerase chain reaction, and flow cytometry analysis were used to elucidate the cytotoxicity of SiNWs. The results showed that the cytotoxicity of SiNWs is greatly dependent on cell lines, SiNWs concentration, and incubation time. Particularly, SiNWs show better biocompatibility with tumor cell lines (eg, human epithelial cervical cancer [Hela] cells and human hepatocellular liver carcinoma [HepG2] cells) than normal cell lines (eg, human normal liver [HL-7702] cells and human embryonic kidney [HEK293T] cells). The reasons may be that SiNWs could tightly attach to the cell membrane in the cell medium, which obviously affects cell adhesion and inhibits their cell viability, especially for normal cell lines. From systematical analysis and comparison, we obtain the concentration limits of SiNWs, which may advance SiNWs applications and its toxicological study in vitro.

4.
Cell Physiol Biochem ; 49(1): 306-321, 2018.
Article in English | MEDLINE | ID: mdl-30138931

ABSTRACT

BACKGROUND/AIMS: To identify new treatment strategies for gastric cancer and to elucidate the mechanism underlying its pathophysiology, we transfected sh-MARCH8 into the human gastric cancer cell lines MKN-45 and AGS to investigate the roles of MARCH8 in gastric cancer. METHODS: We used genetic engineering to construct the sh-MARCH8 interference plasmid and transfected it into gastric cancer cells. Colony formation assays and cell viability measurements were performed to detect the viability and proliferation of cancer cells. Wound healing assays were performed to estimate the migration and proliferation rates of the cells. Cell invasion assays were used to estimate the invasive abilities of the cells. Cell apoptosis analysis was performed by using flowing cytometry. Western blot analysis was performed to estimate the expression levels of proteins. Statistical analysis was performed using the SPSS 18.0 software. Student's t-test was used to determine the significance of all pairwise comparisons of interest. RESULTS: We observed that the transfection of sh-MARCH8 inhibited the survival and proliferation of MKN-45 and AGS cells. The migration and invasion of the MKN-45 and AGS cells were significantly decreased, and apoptosis was induced in comparison with the control cells. These results were further confirmed by data showing that sh-MARCH8 increased the BAX/BCL2 ratio in MKN-45 and AGS cells. We also observed that sh-MARCH8 inactivated the PI3K and ß-catenin stat3 signaling pathways by changing protein expression levels or the phosphorylation of related proteins. CONCLUSION: These data suggested that sh-March8 reduced viability and induced apoptosis of the MKN-45 and AGS cells through the PI3K and ß-catenin stat3 signaling pathways. Taken together, our data revealed that transfection of sh-MARCH8 into the MKN-45 and AGS gastric cancer cell lines inhibited their growth, and this approach may be useful as a novel strategy for gastric cancer therapy.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , RNA, Small Interfering/metabolism , Stomach Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Female , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , STAT3 Transcription Factor/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , bcl-2-Associated X Protein/metabolism , beta Catenin/metabolism
5.
Cell Physiol Biochem ; 44(6): 2158-2173, 2017.
Article in English | MEDLINE | ID: mdl-29241186

ABSTRACT

BACKGROUND/AIMS: MicroRNA-21 is an oncogenic miR (oncomiR) frequently elevated in gastric cancer (GC). Overexpression of miR-21 decreases the sensitivity of GC cells to 5-fluorouridine (5-Fu) and trastuzumab, a humanized monoclonal antibody targeting human epidermal growth factor receptor 2 (HER2). Receptor-mediated endocytosis plays a crucial role in the delivery of biotherapeutics including anti-miRNA oligonucleotides (AMOs). This study is a continuation of earlier findings involving poly(ε-caprolactone) (PCL)-poly (ethylene glycol) (PEG) nanoparticles (PEG-PCL NPs), which were coated with trastuzumab to target GC with HER2 receptor over-expression using anti-miRNA-21 (AMO-21) and 5-Fu. METHODS: HER-PEG-PCL NPs were prepared by one-step carbodiimide coupling using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDAc) and Sulfo-NHS in aqueous phase. Covalent coupling of amino groups at the surface of PEG-PCL with the carboxyl groups of trastuzumab was analyzed by X-ray photoelectron spectroscopy (XPS). AMO-21/5-Fu NPs were formulated by a double-emulsion solvent evaporation technique. The cell line specificity, cellular uptake and AMO-21 delivery were investigated through the rhodamine-B-labeled 6-carboxyfluorescein (FAM)-AMO-21-PEG-PCL NPs coated with or without the antibody in both Her2-positive (NUGC4) and negative GC cells (SGC7901) visualized by fluorescence microscopy. The cytotoxicity of the HER-PEG-PCL NPs encapsulating AMO-21 was evaluated by MTT and apoptosis. Real-time reverse-transcription polymerase chain reaction (RT-PCR) was used to examine miR-21 and phosphatase and tensin homolog (PTEN) and Sprouty2 expression in GC cell lines. The antitumor effects of AMO-21/5-Fu NPs were compared with other groups in xenograft gastric cancer mice. RESULTS: The antibody conjugates significantly enhanced the cellular uptake of NPs. The AMO-21/5-Fu NPs effectively suppressed the target miRNA expression in GC cells, which further up-regulated PTEN and Sprouty2. As a result, the sensitivity of HER2-expressing gastric cancer to trastuzumab and 5-Fu were enhanced both in vitro and in vivo. The approach enhanced the targeting by trastuzumab as well as antibody-dependent cellular cytotoxicity (ADCC) of immune effector cells Conclusions: Taken together, the results provide insight into the biological and clinical potential of targeted AMO-21 and 5-Fu co-delivery using modified trastuzumab for GC treatment.


Subject(s)
Antagomirs/genetics , Antagomirs/therapeutic use , Antineoplastic Agents/therapeutic use , MicroRNAs/genetics , Stomach Neoplasms/therapy , Trastuzumab/therapeutic use , Uridine/analogs & derivatives , Animals , Antagomirs/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Carriers/chemistry , Drug Delivery Systems , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Polyesters/chemistry , Receptor, ErbB-2/analysis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trastuzumab/administration & dosage , Uridine/administration & dosage , Uridine/therapeutic use
6.
Oncotarget ; 8(40): 67189-67202, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978026

ABSTRACT

MiR-21 is an oncogenic miR frequently elevated in gastric cancer. Overexpression of miR-21 decreases the sensitivity of gastric cancer cells to trastuzumab, which is a humanized monoclonal antibody targeting human epidermal growth factor receptor 2. However, optimization of miRNA or its anti-miRNA oligonucleotides (AMOs) for delivery is a challenge. Receptor-mediated endocytosis plays a crucial role in the delivery of biotherapeutics including AMOs. This study is a continuation of our earlier findings involving poly(ε-caprolactone) (PCL)-poly (ethylene glycol) (PEG) nanoparticles (PEG-PCL NPs), which were coated with trastuzumab to target gastric cancer cells with HER2 receptor over-expression using anti-miRNA-21 antisense oligonucleotides (AMO-21). The antibody conjugates (HER-PEG-PCL NPs) act against target cells via antibody-dependent mechanisms and also based on encapsutalated AMO-21. X-ray photoelectron spectroscopy validated the presence of trastuzumab on NP surface. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) revealed a stable antibody expression. The cell line specificity, cellular uptake, AMO-21 delivery, and cytotoxicity of the HER-PEG-PCL NPs were investigated. We found that the antibody conjugates significantly enhanced the cellular uptake of NPs. The HER-PEG-PCL NPs effectively suppressed the target miRNA expression in gastric cancer cells, which further up-regulated phosphatase and tensin homolog (PTEN). As a result, the sensitivity of HER2-expressing gastric cancer cells to trastuzumab was enhanced. The approach enhances the targeting by trastuzumab as well as antibody-dependent cellular cytotoxicity of immune effector cells. The antitumor effects of AMO-21-HER-PEG-PCL NPs were compared with trastuzumab in xenograft gastric cancer mice. The results provide insight into the biological and clinical potential of targeted AMO-21 delivery using modified trastuzumab for gastric cancer treatment.

7.
Oncotarget ; 8(70): 114495-114505, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29383097

ABSTRACT

Hypermethylation of the transcription factor AP-2 epsilon (TFAP2E) gene affects 5-fluorouridine (5-FU) resistance in gastric cancer (GC) patients. The epigenetic inhibitor 5-Aza-2'-deoxycytidine (DAC), which reverses DNA methylation by targeting DNA methyltransferases (DNMTs), has potential to sensitize GC to 5-FU. Nevertheless, DNA demethylation only DAC transiently occurs since DAC is unstable in aqueous solutions, which limits its potential. Here we developed intelligent nanoparticles (NPs) comprising gelatinase with polyethylene glycol (PEG) and poly-ε-caprolactone) (PCL) to specifically deliver DAC (DAC-TNPs) to tumors. DAC-carrying PEG-PCL NPs (DAC-NPs) lacking gelatinase features served as controls. 72 hours after administration of DAC-TNPs or DAC-NPs, 5-FU was sequentially applied to GC cells and human GC xenografts in nude mice. Both in vitro and in vivo evaluations demonstrated that the combination treatment of DAC-TNPs and 5-FU greatly improved tumor suppression in GC cells and mouse xenograft models with hypermethylation TFAP2E (MKN45 cells). We thus propose that the sequential administration of DAC-TNPs and 5-FU could be significant in the development of novel targeted therapies.

8.
Cell Oncol (Dordr) ; 35(3): 175-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22476961

ABSTRACT

AIM: To explore the association between the galectin-3 genetic polymorphisms and Platinum-based chemotherapy response as well as the prognosis of non-small cell lung cancer (NSCLC). METHOD: Three hundred twenty patients with Stage III (A+B) or IV NSCLC were enrolled. A Platinum-based chemotherapy was given to each subjects and therapeutic effect was evaluated. The two galectin-3 genetic polymorphisms, namely, galectin-3 +191 A>C and +292 A>C, were genotyped. RESULTS: The polymorphic genotypes and the allele frequency of galectin-3 +191 A>C were not significantly different between responders and non-responders to chemotherapy. For galectin-3 +292 A>C, the AA genotype and A allele distribution were significantly higher in responders than in non-responders. Logistic regression analysis showed CC genotype of galectin-3 +292 presented higher risk of being non-responders compared with the AA genotype (OR = 2.96, 95 % CI: 1.55-5.47; P < 0.001). The overall survival in patients with AA genotype of galectin-3 +292 were significantly longer than in those with CC genotype (25.6 vs. 19.5 months, P = 0.013). The hazard ratio for CC genotype of galectin-3 +292 was 2.43 (95 % CI: 2.03-3.98, compared with AA carriers, P = 0.003). CONCLUSION: The galectin-3 genetic polymorphisms of galectin-3 +292, rather than galectin-3 +191, were associated with the chemotherapy response and prognosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Galectin 3/genetics , Genetic Predisposition to Disease , Lung Neoplasms/drug therapy , Platinum/therapeutic use , Polymorphism, Single Nucleotide/genetics , Adult , Aged , Female , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Prognosis , Proportional Hazards Models , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...