Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Language
Publication year range
1.
Braz J Med Biol Res ; 52(4): e7728, 2019.
Article in English | MEDLINE | ID: mdl-30994730

ABSTRACT

Pituitary adenoma is one of the most common tumors in the neuroendocrine system. This study investigated the effects of long non-coding RNAs (lncRNAs) highly up-regulated in liver cancer (HULC) on rat secreting pituitary adenoma GH3 cell viability, migration, invasion, apoptosis, and hormone secretion, as well as the underlying potential mechanisms. Cell transfection and qRT-PCR were used to change and measure the expression levels of HULC, miR-130b, and FOXM1. Cell viability, migration, invasion, and apoptosis were assessed using trypan blue staining assay, MTT assay, two-chamber transwell assay, Guava Nexin assay, and western blotting. The concentrations of prolactin (PRL) and growth hormone (GH) in culture supernatant of GH3 cells were assessed using ELISA. The targeting relationship between miR-130b and FOXM1 was verified using dual luciferase activity. Finally, the expression levels of key factors involved in PI3K/AKT/mTOR and JAK1/STAT3 pathways were evaluated using western blotting. We found that HULC was highly expressed in GH3 cells. Overexpression of HULC promoted GH3 cell viability, migration, invasion, PRL and GH secretion, as well as activated PI3K/AKT/mTOR and JAK1/STAT3 pathways. Knockdown of HULC had opposite effects and induced cell apoptosis. HULC negatively regulated the expression of miR-130b, and miR-130b participated in the effects of HULC on GH3 cells. FOXM1 was a target gene of miR-130b, which was involved in the regulation of GH3 cell viability, migration, invasion, and apoptosis, as well as PI3K/AKT/mTOR and JAK1/STAT3 pathways. In conclusion, HULC tumor-promoting roles in secreting pituitary adenoma might be via down-regulating miR-130b, up-regulating FOXM1, and activating PI3K/AKT/mTOR and JAK1/STAT3 pathways.


Subject(s)
Adenoma/pathology , Pituitary Neoplasms/pathology , RNA, Long Noncoding/physiology , Adenoma/genetics , Adenoma/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Migration Assays , Cell Movement/physiology , Cell Survival/physiology , Enzyme-Linked Immunosorbent Assay , Forkhead Box Protein M1/analysis , Forkhead Box Protein M1/metabolism , Humans , Janus Kinase 1/analysis , Janus Kinase 1/metabolism , Luciferases , MicroRNAs/analysis , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/analysis , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/analysis , STAT3 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/analysis , TOR Serine-Threonine Kinases/metabolism , Transfection
2.
Braz. j. med. biol. res ; 52(4): e7728, 2019. graf
Article in English | LILACS | ID: biblio-1001506

ABSTRACT

Pituitary adenoma is one of the most common tumors in the neuroendocrine system. This study investigated the effects of long non-coding RNAs (lncRNAs) highly up-regulated in liver cancer (HULC) on rat secreting pituitary adenoma GH3 cell viability, migration, invasion, apoptosis, and hormone secretion, as well as the underlying potential mechanisms. Cell transfection and qRT-PCR were used to change and measure the expression levels of HULC, miR-130b, and FOXM1. Cell viability, migration, invasion, and apoptosis were assessed using trypan blue staining assay, MTT assay, two-chamber transwell assay, Guava Nexin assay, and western blotting. The concentrations of prolactin (PRL) and growth hormone (GH) in culture supernatant of GH3 cells were assessed using ELISA. The targeting relationship between miR-130b and FOXM1 was verified using dual luciferase activity. Finally, the expression levels of key factors involved in PI3K/AKT/mTOR and JAK1/STAT3 pathways were evaluated using western blotting. We found that HULC was highly expressed in GH3 cells. Overexpression of HULC promoted GH3 cell viability, migration, invasion, PRL and GH secretion, as well as activated PI3K/AKT/mTOR and JAK1/STAT3 pathways. Knockdown of HULC had opposite effects and induced cell apoptosis. HULC negatively regulated the expression of miR-130b, and miR-130b participated in the effects of HULC on GH3 cells. FOXM1 was a target gene of miR-130b, which was involved in the regulation of GH3 cell viability, migration, invasion, and apoptosis, as well as PI3K/AKT/mTOR and JAK1/STAT3 pathways. In conclusion, HULC tumor-promoting roles in secreting pituitary adenoma might be via down-regulating miR-130b, up-regulating FOXM1, and activating PI3K/AKT/mTOR and JAK1/STAT3 pathways.


Subject(s)
Humans , Animals , Rats , Pituitary Neoplasms/pathology , Adenoma/pathology , RNA, Long Noncoding/physiology , Enzyme-Linked Immunosorbent Assay , Transfection , Adenoma/genetics , Adenoma/metabolism , Cell Movement/physiology , Cell Survival/physiology , Blotting, Western , Apoptosis/physiology , MicroRNAs/analysis , Cell Line, Tumor , STAT3 Transcription Factor/analysis , Janus Kinase 1/analysis , Janus Kinase 1/metabolism , Cell Migration Assays , Forkhead Box Protein M1/analysis , Forkhead Box Protein M1/metabolism , Luciferases
3.
Pharmazie ; 73(7): 402-407, 2018 07 01.
Article in English | MEDLINE | ID: mdl-30001775

ABSTRACT

OBJECTIVE: Curcumin has been reported to possess anti-tumor effects on multiple cancers, including lung cancer. However, the mechanisms of its anti-tumor effect on lung cancer have not been fully elucidated. Our study attempted to identify the effect of curcumin on A549 cells and further explore the potential mechanism. METHODS: Different concentrations of curcumin were exposed to A549 cells for 24 h and cell viability was measured by CCK-8 assay. The expression of UCA1 was overexpressed in A549 cells by transfection with pEX-UCA1. Cell proliferation was determined by BrdU staining and assessing the expression of CyclinD1 using western blot and RT-PCR assay. Apoptotic cells were measured by flow cytometry assay. Western blot was performed to assess the expression of apoptosis-related, Wnt and mTOR pathways-related factors. RESULTS: Curcumin incubation dramatically reduced viability of A549 cells in a dosage-dependent manner. Curcumin (0.6 µM) significantly reduced BrdU+-positive cells, declined the expression of CyclinD1, and enhanced cell apoptosis. Interestingly, we found that curcumin inhibited the expression of UCA1 and UCA1 overexpression abolished the effect of curcumin on cell apoptosis. In addition, we also found that curcumin inhibited Wnt and mTOR pathways through down-regulation of UCA1. CONCLUSION: We demonstrated that curcumin inhibited the growth of A549 cells through downregulation of UCA1, which might provide new insight for the treatment of lung cancer.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Lung Neoplasms/drug therapy , RNA, Long Noncoding/genetics , A549 Cells , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/administration & dosage , Dose-Response Relationship, Drug , Down-Regulation , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/drug effects
4.
Int J Clin Exp Pathol ; 11(7): 3257-3266, 2018.
Article in English | MEDLINE | ID: mdl-31949700

ABSTRACT

Background: Myocardial infarction (MI) is a serious condition, caused by acute, persistent ischemia or hypoxia of a coronary artery and responsible for heart failure and sudden death. This study aimed to investigate the effects of catechin, one of the main active components of green tea, on hypoxia-induced MI cell model, as well as the underlying possible mechanism. Methods: Cell viability, proliferation, apoptosis, and the expression of microRNA-92a (miR-92a) after hypoxia stimulation and/or catechin treatment were assessed using cell counting kit-8 (CCK-8) assay, western blotting, annexin V-FITC/PI staining and qRT-PCR, respectively. miRNA transfection was performed to change the expression of miR-92a. The effects of miR-92a on hypoxia and catechin-treated H9c2 cell viability, proliferation and apoptosis were evaluated. Finally, western blotting was conducted to measure the expression of core factors in the c-Jun N-terminal kinase (JNK) signaling pathway. Results: Hypoxia stimulation significantly inhibited H9c2 cell viability and proliferation, induced cell apoptosis and up-regulated miR-92a expression. Catechin markedly protected H9c2 cells from hypoxia-induced viability loss, proliferation inhibition, and apoptosis enhance, as well as miR-92a expression increase. Furthermore, suppression of miR-92a enhanced the protective effects of catechin on hypoxia-induced H9c2 cells. Overexpression of miR-92a had opposite effects. Catechin activated the JNK pathway in H9c2 cells by down-regulating miR-92a. Conclusion: Catechin protected H9c2 cells from hypoxia-induced injury by regulating miR-92a and JNK signaling pathway. Our findings facilitate the understanding of the protective activity of catechin in hypoxia-induced MI cell injury and provide a theoretical basis for further explore treatment of MI by using catechin.

5.
Int J Clin Exp Pathol ; 11(8): 3876-3883, 2018.
Article in English | MEDLINE | ID: mdl-31949775

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is a common tumor of bone, and the high incidence and poor prognosis of OS call for novel therapeutic strategies. We aimed to explore the functional role of lentinan (LNT) in human OS MG63 cells as well as the underlying mechanisms. METHODS: Cell viability of MG63 cells under LNT stimulation was measured by CCK-8 assay to explore the adequate concentration of LNT. Cell proliferation, apoptosis and expression of microRNA (miR)-340 in MG63 cells after LNT treatments were assayed by BrdU incorporation assay, flow cytometry assay and quantitative reverse transcription PCR, respectively. Expression of proteins associated with cell cycle, apoptosis, and autophagy were determined by western blot analysis. Subsequently, whether LNT affected MG63 cells through miR-340 as well as the related signaling pathway was explored. RESULTS: Cell viability was reduced by 5-100 mg/mL of LNT. Percentage of BrdU-positive cells was reduced while that of apoptotic cells was enhanced by LNT treatment. LNT decreased cyclin D1 level but increased levels of active caspase-3 and caspase-9. After treatment, LNT enhanced LC3B-II/LC3B-I and Beclin-1 levels but reduced the p62 level. The miR-340 level was up-regulated by LNT, and further experiments showed LNT promoted apoptosis and autophagy through up-regulating miR-340. Moreover, LNT reduced the phosphorylated levels of MAPK and ERK through up-regulating miR-340. CONCLUSION: LNT reduced proliferation and induced apoptosis and autophagy by up-regulating miR-340 in MG63 cells, along with inhibition of the MAPK/ERK pathway.

6.
Asian Pac J Cancer Prev ; 15(15): 6269-73, 2014.
Article in English | MEDLINE | ID: mdl-25124610

ABSTRACT

Growing evidence suggests that miR-150-5p has an important role in regulating genesis of various types of cancer. However, the roles and the underlying mechanisms of miR-150-5p in development of colorectal cancer (CRC) remain largely unknown. Transwell chambers were used to analyze effects on cell migration and invasion by miR-150-5p. Quantitative real-time PCR (qRT-PCR), Western blotting and dual-luciferase 3' UTR reporter assay were carried out to identify the target genes of miR-150-5p. In our research, miR-150-5p suppressed CRC cell migration and invasion, and MUC4 was identified as a direct target gene. Its effects were partly blocked by re-expression of MUC4. In conclusiomn, miR-150-5p may suppress CRC metastasis through directly targeting MUC4, highlighting its potential as a novel agent for the treatment of CRC metastasis.


Subject(s)
Cell Movement , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Mucin-4/metabolism , 3' Untranslated Regions/genetics , Blotting, Western , Cell Proliferation , Colorectal Neoplasms/metabolism , Humans , Luciferases/metabolism , Mucin-4/genetics , Neoplasm Invasiveness , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...