Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biochem Biophys Res Commun ; 576: 108-116, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34482023

RESUMO

Ras-related GTP binding (Rag) GTPases are required to activate mechanistic target of rapamycin complex 1 (mTORC1), which plays a central role in cell growth and metabolism and is considered as one of the most important oncogenic pathways. Therefore, Rag GTPases have been speculated to play a pro-cancer role via mTOR induction. However, aside from stimulation of mTOR signaling, firm links connecting Rag GTPase activity and their downstream effectors with cancer progression, remain largely unreported. In this study, we reported a novel link between RagB/C and a known oncoprotein phosphatase of regenerating liver-3 (PRL-3) by screening 22 pairs of tumors and their adjacent normal tissues from gastric, liver and lung cancers, and validating our findings in cancer cell lines with ectopic RagB/C expression. RagB/C was found to enhance PRL-3 stability by modulating two major cellular protein degradation pathways: lysosomal-autophagy and ubiquitin-proteasome system (UPS). Functionally, we identified the correlation between RagB/C expression with poor clinical outcomes in breast or colon cancer patients who also showed low PRL-3 mRNA expression from data retrieved from TCGA datasets, highlighting the potential relevance of Rag GTPase and PRL-3 mRNA in combination as a prognostic clinical biomarker.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Linhagem Celular Tumoral , Biologia Computacional , Bases de Dados Genéticas , Humanos , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Prognóstico , Ligação Proteica , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteólise , Transdução de Sinais , Taxa de Sobrevida
2.
J Cell Biochem ; 120(9): 15941-15951, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31099097

RESUMO

BACKGROUND: HOMEOBOX A11 (HOXA11) antisense RNA (HOXA11-AS), a newly identified long noncoding RNAs, is involved in the carcinogenic process of several human tumors. However, the role of HOXA11-AS in liver cancer progression is not well understood. MATERIALS AND METHODS: This study used liver cancer tissues and cell lines (CSQT-2 and HCCLM3) to explore the potential mechanism of HOXA11-AS. Quantitative real-time polymerase chain reaction and Western blot were applied to evaluate the bio-molecules expression. Bioinformatics analysis, RNA pull down and luciferase report assay were applied to determine the molecules bind. MTT, transwell, and wound healing assay were used to measure the cell growth situation. RESULTS: HOXA11-AS was highly expressed in liver cancer tissues and cell lines, which was closely related with poor prognosis in patients with liver cancer. HOXA11-AS could act as a competing endogenous RNA for miR-15a-3p. Besides, miR-15a-3p negatively controlled its target molecule signal transducer and activator of transcription 3 (STAT3). Furthermore, a linear regression analysis and biological experiments showed a positive correlation between HOAX11-AS and STAT3. Moreover, HOAX11-AS overexpression activated the Janus kinase (JAK)-STAT pathway and thus promoted the growth, migration, and invasion of liver cancer cells, which might be through regulating miR-15a-3p/STAT3 axis. CONCLUSION: Our study suggested that HOAX11-AS could regulate the JAK-STAT signaling pathway by miR-15a-3p/STAT3 axis to promote the progression of liver cancer. Thus, HOXA11-AS may be regarded as an effective biomarker with diagnostic, prognostic, and therapeutic potentials for liver cancer.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , MicroRNAs/genética , RNA Longo não Codificante/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Janus Quinases/metabolismo , Neoplasias Hepáticas/genética , Masculino , Metástase Neoplásica , Estadiamento de Neoplasias , Prognóstico , Transdução de Sinais , Carga Tumoral
3.
Biochem Biophys Res Commun ; 482(4): 597-603, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27864146

RESUMO

Aberrant expression of microRNAs contributes to the initiation and progression of numerous human cancers. The underlying effects and molecular mechanisms of microRNA-187 (miR-187) in gastric cancer (GC) remain unclear. The present study reports that miR-187 was significantly overexpressed in GC tissues compared to that in non-tumor tissues and was associated with malignant clinical factors such as depth of invasion (P = 0.005), tumor size (P = 0.024), lymph node metastasis (P = 0.048), and TNM stage (P = 0.035). Additionally, miR-187 promoted tumor growth in vivo, and significantly increased migration, invasion, and proliferation, but inhibited apoptosis in GC cells. It was found that collapsin response mediator protein 1 (CRMP1), a tumor suppressor, was a direct downstream target of miR-187 in GC. Furthermore, CRMP1 silencing resulted in similar effects on cell proliferation, migration, and apoptosis as those of miR-187 overexpressing GC cells. Additionally, the effects of miR-187 inhibitor on cell migration and cell apoptosis were reversed by CRMP1 downregulation. In summary, miR-187 promotes tumor progression by regulating CRMP1 expression in GC and may thus be a potential prognostic marker and a therapeutic target in GC.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Fosfoproteínas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Estômago/patologia , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia
4.
Oncogenesis ; 13(1): 16, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769340

RESUMO

Accumulating studies have shown that E3 ligases play crucial roles in regulating cellular biological processes and signaling pathways during carcinogenesis via ubiquitination. Tripartite-motif (TRIM) ubiquitin E3 ligases consist of over 70 members. However, the clinical significance and their contributions to tumorigenesis remain largely unknown. In this study, we analyzed the RNA-sequencing expression of TRIM E3 ligases in colorectal cancer (CRC) and identified 10 differentially expressed genes, among which TRIM1 expression predicted poor prognosis of CRC patients. We demonstrated that TRIM1 expression is positively associated with CRC pathological stages, and higher expression is positively correlated with infiltrating levels of immune cells and immunotherapy biomarkers. TRIM1 expression promotes the proliferation and migration of colorectal cancer cells in vitro and in vivo. Transcriptional analysis showed that TRIM1 is responsible for metabolism promotion and immune suppression. Mechanistically, we found that TRIM1 binds HIF1α and mediates its K63-linked ubiquitination, which is required for HIF1α nuclear translocation and subsequent activation. Ubiquitination occurs at Lys214 in the loop between the two PAS domains of HIF1α, and mutation of Lys214 severely disturbs the function of HIF1α. Besides, HIF1α ubiquitination enhances its binding with proteins involved in cellular trafficking and nucleocytoplasmic transport pathway. Collectively, our results indicate TRIM1's role in predicting prognosis and reveal how TRIM1 functions to upregulate HIF1α expression and promote tumor cell proliferation.

5.
Sci Rep ; 14(1): 7733, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565963

RESUMO

B-Myb has received considerable attention for its critical tumorigenic function of supporting DNA repair. However, its modulatory effects on chemotherapy and immunotherapy have rarely been reported in colorectal cancer. Bortezomib (BTZ) is a novel compound with chemotherapeutic and immunotherapeutic effects, but it fails to work in colorectal cancer with high B-Myb expression. The present study was designed to investigate whether B-Myb deletion in colorectal cancer could potentiate the immune efficacy of BTZ against colorectal cancer and to clarify the underlying mechanism. Stable B-Myb knockdown was induced in colorectal cancer cells, which increased apoptosis of the cancer cells relative to the control group in vitro and in vivo. We found that BTZ exhibited more favourable efficacy in B-Myb-defective colorectal cancer cells and tumor-bearing mice. BTZ treatment led to differential expression of genes enriched in the p53 signaling pathway promoted more powerful downstream DNA damage, and arrested cell cycle in B-Myb-defective colorectal cancer. In contrast, recovery of B-Myb in B-Myb-defective colorectal cancer cells abated BTZ-related DNA damage, cell cycle arrest, and anticancer efficacy. Moreover, BTZ promoted DNA damage-associated enhancement of immunogenicity, as indicated by potentiated expression of HMGB1 and HSP90 in B-Myb-defective cells, thereby driving M1 polarization of macrophages. Collectively, B-Myb deletion in colorectal cancer facilitates the immunogenic death of cancer cells, thereby further promoting the immune efficacy of BTZ by amplifying DNA damage. The present work provides an effective molecular target for colorectal cancer immunotherapy with BTZ.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Animais , Camundongos , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Morte Celular Imunogênica , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Apoptose
6.
Photodiagnosis Photodyn Ther ; 42: 103558, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37030434

RESUMO

OBJECTIVE: Photodynamic therapy (PDT) may be an effective therapeutic strategy for colorectal cancer at an early stage. However, malignant cells' resistance to photodynamic agents can lead to treatment failure. MYBL2 (B-Myb) is an oncogene in colorectal carcinogenesis and development, for which little research has focused on its effect on drug resistance. MATERIALS AND METHODS: In the present work, a colorectal cancer cell line with a stable knockdown of MYBL2 (ShB-Myb) was constructed first. Chlorin e6 (Ce6) was utilized to induced PDT. The anti-cancer efficacy was measured by CCK-8, PI staining, and Western blots. The drug uptake of Ce6 was assayed by flow cytometry and confocal microscopy. The ROS generation was detected by the CellROX probe. DDSB and DNA damage were assayed through comet experiment and Western blots. The over-expression of MYBL2 was conducted by MYBL2 plasmid. RESULTS: The findings indicated that the viability of ShB-Myb treated with Ce6-PDT was not decreased compared to control SW480 cells (ShNC), which were resistant to PDT. Further investigation revealed reduced photosensitizer enrichment and mitigated oxidative DNA damage in colorectal cancer cells with depressed MYBL2. It turned out that SW480 cells knocking down MYBL2 showed phosphorylation of NF-κB and led to up-regulation of ABCG2 expression thereupon. When MYBL2 was replenished back in MYBL2-deficient colorectal cancer cells, phosphorylation of NF-κB was blocked and ABCG2 expression up-regulation was suppressed. Additionally, replenishment of MYBL2 also increased the enrichment of Ce6 and the efficacy of PDT. CONCLUSION: In summary, MYBL2 absence in colorectal cancer contributes to drug resistance by activating NF-κB to up-regulate ABCG2 and thereby leading to photosensitizer Ce6 efflux. This study provides a novel theoretical basis and strategy for how to effectively improve the anti-tumor efficacy of PDT.


Assuntos
Clorofilídeos , Neoplasias Colorretais , Fotoquimioterapia , Porfirinas , Humanos , Fármacos Fotossensibilizantes/farmacologia , Fotoquimioterapia/métodos , Regulação para Cima , NF-kappa B/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Porfirinas/farmacologia , Linhagem Celular Tumoral , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Proteínas de Neoplasias , Transativadores/metabolismo , Proteínas de Ciclo Celular/metabolismo
7.
Front Pharmacol ; 13: 854526, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35662735

RESUMO

Aloe-emodin (1,8-dihydroxy-3-hydroxymethyl-anthraquinone), derived from some Chinese edible medicinal herbs, exerts a potential anticancer activity on various cancer cells, making it a drug candidate for cancer therapy. Yet, the role of aloe-emodin in pyroptosis, a new type of cell death, is uncharacterized. In this study, we explored the molecular mechanisms of aloe-emodin-triggered pyroptosis. Aloe-emodin inhibited proliferation and migration and triggered caspase-dependent cell death of HeLa cells in a dose-dependent manner. Aloe-emodin caused mitochondrial dysfunction and induced pyroptosis by activating the caspase-9/3/GSDME axis. Transcriptional analysis showed extensive changes in gene expressions in cellular pathways, including MAPK, p53, and PI3K-Akt pathways when treated with aloe-emodin. This study not only identified a novel role of aloe-emodin in pyroptotic cell death, but also performed a systematical genome-wide analysis of cellular pathways responding to aloe-emodin, providing a theoretical basis for applying anthraquinone derivatives in the treatment of GSDME-expressing cancers.

8.
J BUON ; 26(4): 1695, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34565051

RESUMO

Retraction of "MicroRNA-23 regulates the growth, metastasis and chemosensitivity of human gastric cancer cells by targeting MAP3K9", by Deping Li, Juan Wang, Yuanjian Hui, Yan Yang, Xingchun Peng, Shengbao Li, Chuantao Sun. JBUON 2020;25(5):2315-2321; PMID: 33277851 Following the publication of the above article, readers drew to our attention that part of the data was unreliable: Figures of this article appeared in other articles (by totally different authors). The authors were requested to provide the raw data and were also asked for an explanation to account for these concerns, but the Editorial Office did not receive any reply. Given above, we decided to retract this article. Authors were informed of the retraction. We thank the readers for bringing this matter to our attention. We apologize for any inconvenience it may cause.

9.
Cell Transplant ; 29: 963689720925059, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32638620

RESUMO

Gastric cancer (GC) is a leading cause of cancer-related death with poor prognosis. Growing evidence has shown that long noncoding ribonucleic acid (lncRNA) FEZ family zinc finger 1 antisense RNA 1(FEZF1-AS1), an "oncogene," regulates tumor progression and supports cancer stem cell. However, the tumorigenic mechanism of FEZF1-AS1 on gastric cancer stem cell (GCSC) is yet to be investigated. Here, we discovered that FEZF1-AS1 was upregulated in GC tissues and cell lines. Knockdown of FEZF1-AS1 inhibited sphere formation and decreased expression of stem factors and markers. Moreover, FEZF1-AS1 silence also suppressed cell proliferation, viability, invasion, and migration of GCSCs. MiR-363-3p is used as a target of FEZF1-AS1, because its expression was suppressed by FEZF1-AS1 in GCSCs. FEZF1-AS1 could sponge miR-363-3p and increased the expression of high-mobility group AT-hook 2 (HMGA2). The expression of FEZF1-AS1 and miR-363-3p, as well as that of miR-363-3p and HMGA2, was negatively correlated in GC tissues. Finally, FEZF1-AS1 contributed to promotion of GCSCs progression partially through inhibition of miR-363-3p. Subcutaneous xenotransplanted tumor model revealed that silence of FEZF1-AS1 suppressed in vivo tumorigenic ability of GSCS via downregulation of HMGA2. In general, our findings clarified the critical regulatory role of FEZF1-AS1/miR-363-3p/HMGA2 axis in GCSC progression, providing a potential therapeutic target for GC.


Assuntos
Proteína HMGA2/metabolismo , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas Repressoras/genética , Neoplasias Gástricas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Feminino , Proteína HMGA2/genética , Humanos , Masculino , Camundongos , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , RNA Antissenso/genética , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Transfecção
10.
J BUON ; 25(5): 2315-2321, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33277851

RESUMO

PURPOSE: The efficient therapeutic targets which could be utilized for gastric cancer are limited. In this context, this study was undertaken to evaluate possible anticancer activity of miR-23 against gastric cancer. METHODS: The normal GES-1 and human AGS, SNU-1 and SNU-5 gastric cancer cells were used in this study. qRT-PCR was used for the determination of miR-23 expression. MTT assay was used for cell viability and acridine orange (AO)/ethidium bromide (EB) assay was used for detection of apoptosis. The experiments were performed in triplicate. RESULTS: The microRNA (miR)-23 was downregulated in gastric cancer cells and showed inhibitory effect on cell growth which was manifested as decline in cell survival. Additionally, the chemosensitivity of gastric cancer cells to cisplatin was enhanced with miR-23 overexpression. Furthermore, miR-23 also inhibited the migration and invasion of cancer cells. MAP3K9 was shown to be the target gene of miR-23 and silencing of MAP3K9 was seen to mimic the growth inhibitory effect of miR-23. Overexpression of MAP3K9 reversed the growth inhibition in miR-23 mimics transfected gastric cancer cells. CONCLUSION: miR-23 exerts growth inhibitory effect against gastric cancer cells and negatively regulates the cell migration and invasion along with enhancement of chemosensitivity of cancer cells.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , MicroRNAs/metabolismo , Neoplasias Gástricas/genética , Apoptose , Movimento Celular , Proliferação de Células , Humanos , Metástase Neoplásica , Neoplasias Gástricas/patologia
11.
Pathol Res Pract ; 210(12): 1011-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24947411

RESUMO

These studies investigated the appearance and function of adrenal glands in rats with severe acute pancreatitis (SAP) and established a new histopathological score to evaluate adrenal histopathological changes. Severe acute pancreatitis relied on retrograde infusion of 5% sodium taurocholate into the bile-pancreatic duct. The damage of SAP was estimated by serum amylase, secretory phospholipase A2 and pancreatic histopathology. Light and electron microscopy of adrenal gland, and the levels of serum corticosterone were investigated. These results showed that the generally ascending trend of adrenal pathological score was inversely proportional to the generally descending trend of serum corticosterone levels, but parallel with the changes of pancreatic histopathology. Herein, the new adrenal histopathological score was effective in the evaluation of adrenal injury following SAP. It may indirectly reflect the variation of serum cortisol levels and the severity of pancreatitis to a certain extent.


Assuntos
Glândulas Suprarrenais/patologia , Pâncreas/patologia , Pancreatite/patologia , Amilases/sangue , Animais , Modelos Animais de Doenças , Masculino , Pancreatite/sangue , Fosfolipases A2 Secretórias/sangue , Ratos , Ratos Wistar , Projetos de Pesquisa , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA