Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Toxicol Res (Camb) ; 13(2): tfae040, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38500512

RESUMO

Objectives: Gastric cancer (GC) is the leading digestive malignancy with high incidence and mortality rate. microRNAs (miRs) play an important role in GC progresssion. This study aimed to investigate the effect of miR-98-5p on proliferation, migration, and invasion of GC cells. Methods: The expression levels of miR-98-5p, ubiquitin specific peptidase 44 (USP44), and CCCTCbinding factor-like (CTCFL) in GC tissues and cells were identified using reversetranscription quantitative polymerase chain reaction and Western blot assay. The relationship between miR-98-5p expression/USP44 and the clinicopathological features in GC patients was analyzed. GC cell proliferation, invasion, and migration were evaluated by cell counting kit-8 and clone formation assays and Transwell assays. The bindings of miR-98-5p to USP44 and USP44 to CTCFL were examined using dualluciferase assay and co-immunoprecipitation. GC cells were treated with MG132 and the ubiquitination level of CTCFL was examined using ubiquitination assay. Rescue experiments were performed to verify the roles of USP44 and CTCFL in GC cells. Results: miR-98-5p was downregulated in GC. miR-98-5p overexpression inhibited the proliferation, migration, and invasion of GC cells. miR-98-5p inhibited USP44 expression. USP44 bound to CTCFL and limited ubiquitination degradation of CTCFL. Overexpression of USP44 and CTCFL attenuated the inhibitory effects of miR-98-5p overexpression on GC cell progression. Conclusion: miR-98-5p overexpression limited USP44-mediated CTCFL deubiquitination, and suppressed CTCFL expression, mitigating GC cell proliferation, migration, and invasion.

2.
Am Surg ; 89(12): 5480-5486, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36787579

RESUMO

OBJECTIVE: This study aimed to investigate the feasibility of the minimal proximal resection margin (PRM) in total gastrectomy (TG) for Siewert II adenocarcinoma of the esophagogastric junction (AEG). METHODS: This study finally included 178 Siewert II advanced AEG patients who underwent TG from January 2017 to September 2020. According to the PRM length, patients were divided into 20-25 mm group and 30-35 mm group. Intraoperative, short-, and long-term postoperative outcomes were compared between two groups. RESULTS: The PRM of the 20-25 mm group had significantly less operation time compared with the PRM of the 30-35 mm group (P < .001), but the amount of blood loss, management of the diaphragmatic crura, and the incidence of positive resection margin were not significantly different between two groups (P > .05). In short-term postoperative outcomes, first gas-passing time, gastric-tube removal time, start time of diet, hospitalization, postoperative complications, and body weight loss were similar between two groups (P > .05). During the follow-up, the 3-year overall survival rates and the recurrence rates were not significantly different between the PRM of 20-25 mm and 30-35 mm groups (81.2% vs 83.5%, P = .695; 18.8% vs 15.5%, P = .812, respectively). CONCLUSION: With less operation time and more preserved esophagus, the minimal PRM length of 20-25 mm could be an appropriate option in TG for patients with Siewert II advanced AEG.


Assuntos
Adenocarcinoma , Neoplasias Esofágicas , Neoplasias Gástricas , Humanos , Estudos Retrospectivos , Margens de Excisão , Junção Esofagogástrica/cirurgia , Junção Esofagogástrica/patologia , Adenocarcinoma/patologia , Gastrectomia , Neoplasias Gástricas/patologia , Neoplasias Esofágicas/cirurgia , Neoplasias Esofágicas/patologia
3.
Int Immunopharmacol ; 115: 109671, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36621327

RESUMO

Acute lung injury (ALI) is characterized by acute systemic inflammatory responses that may lead to severe acute respiratory distress syndrome (ARDS). The clinical course of ALI/ARDS is variable; however, it has been reported that lipopolysaccharides (LPS) play a role in its development. The fragile chromosomal site gene WWOX is highly sensitive to genotoxic stress induced by environmental exposure and is an important candidate gene for exposure-related lung disease research. However, the expression of WWOX and its role in LPS-induced ALI still remain unidentified. This study investigated the expression of WWOX in mouse lung and epithelial cells and explored the role of WWOX in LPS-induced ALI model in vitro and in vivo. In addition, we explored one of the possible mechanisms by which WWOX alleviates ALI from the perspective of autophagy. Here, we observed that LPS stimulation reduced the expression of WWOX and the autophagy marker microtubule-associated protein 1 light chain 3ß-II (MAP1LC3B/LC3B) in mouse lung epithelial and human epithelial (H292) cells. Overexpression of WWOX led to the activation of autophagy and inhibited inflammatory responses in LPS-induced ALI cells and mouse model. More importantly, we found that WWOX interacts with mechanistic target of rapamycin [serine/threonine kinase] (mTOR) and regulates mTOR and ULK-1 signaling-mediated autophagy. Thus, reduced WWOX levels were associated with LPS-induced ALI. WWOX can activate autophagy in lung epithelial cells and protect against LPS-induced ALI, which is partly related to the mTOR-ULK1 signaling pathway.


Assuntos
Lesão Pulmonar Aguda , Síndrome do Desconforto Respiratório , Camundongos , Animais , Humanos , Lipopolissacarídeos/toxicidade , Serina-Treonina Quinases TOR/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Pulmão/metabolismo , Inflamação/metabolismo , Síndrome do Desconforto Respiratório/metabolismo , Autofagia , Oxidorredutase com Domínios WW/genética , Oxidorredutase com Domínios WW/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
4.
Cell Mol Biol (Noisy-le-grand) ; 69(14): 143-149, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38279460

RESUMO

This study aimed to investigate the possible mechanism of Micro RNA-548b-5p (miR-548b-5p) down-regulating frizzled (FZD) 7 to suppress the migration and invasion of gastric carcinoma cells. For this purpose, HGCC (Human gastric carcinoma cell) lines were selected (Hs-746T, NCI-N87, SGC-7901, MKN-45, SNU-1), and human normal gastric mucosa cells GES-1. QRT PCR was adopted to reveal and screen the cell line with low expression of mir-548b-5p (hs-746t) for research; the Hs-746T cells were randomly assigned into control group, miR-548b-5p NC group, miR-548b-5p mimic group, miR-548b-5p mimic+pc-FZD7 group. The CCK-8 assay was utilized to measure Hs-746T cell viability, while flow cytometry, Trans well chamber, and scratch test were utilized to examine the apoptotic, invasive, and migratory properties of the cells, respectively. WB was used to detect the SATB1, as well as the expression levels of proteins involved in apoptosis, including Caspase-3, Bax, and Bcl-2, as well as Matrix metalloproteinase 2 and 9 (MMP-2 and MMP-9) in SW620 cells. The binding of miR-548b-5p to FZD7 was evaluated through the dual-luciferase reporter assay. The results indicate that MiR-548b-5p showed low expression in HGCCs; in contrast to the control group (P>0.05), the Hs-746T cell viability, invasion, migration ability, MMP-2, MMP-9 protein significantly downregulated in miR-548b-5p mimic group (P<0.05), the apoptosis rate, Caspase-3, Bax protein expression were upregulated markedly, and Bcl-2 protein expression was downregulated significantly (P<0.05); in contrast to miR-548b-5p mimic group, the Hs-746T cell viability, invasion, migration ability, MMP-2, MMP-9 protein significantly were upregulated in miR-548b-5p mimic+pc-FZD7 group (P<0.05), the apoptosis rate, Caspase-3, Bax protein expression were significantly, and the level of Bcl-2 was down-regulated significantly (P<0.05); Double Luciferase Report shows that mir-548b-5p can target and regulate fzd7. It was concluded that MiR-548b-5p can suppress cell growth and migration of HGCC Hs-746T, which may be achieved by targeted down-regulation of FZD7.


Assuntos
Carcinoma , Receptores Frizzled , Proteínas de Ligação à Região de Interação com a Matriz , MicroRNAs , Humanos , Apoptose/genética , Proteína X Associada a bcl-2/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Luciferases/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
5.
Front Pharmacol ; 13: 994625, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518680

RESUMO

Acute lung injury (ALI) is a severe clinical disorder characterized by dysregulated inflammatory responses, leading to high rates of morbidity and mortality. Cinobufagin, a primary component isolated from cinobufotalin, exerts strong anticancer effects. However, there are few reports on its role in ALI, and it is unclear whether cinobufagin affects lipopolysaccharide (LPS)-induced ALI. Therefore, the present study aimed to investigate the effect of cinobufagin on LPS-induced ALI and to assess its potential mechanism of action. The results showed that cinobufagin alleviated lung histopathological changes and protected the permeability of lung tissues in LPS-induced ALI. In addition, cinobufagin effectively suppressed inflammatory responses through the induction of autophagy in LPS-induced ALI cells and in a mouse model. Moreover, cinobufagin enhanced autophagy through the p53/mTOR pathway in LPS-induced ALI. Herein, it was reported for the first time that cinobufagin inhibited the inflammatory response of LPS-induced ALI, which laid the foundation for further understanding and development of cinobufagin as a potential new drug for ALI.

6.
J Healthc Eng ; 2022: 5390182, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35719717

RESUMO

To explore the clinical effects of total laparoscopic radical gastrectomy under the guidance of the concept of enhanced recovery after surgery (ERAS). Fifty-five patients were perioperatively treated under the concept of ERAS (ERAS group), while the remaining 55 patients were treated under the traditional perioperative concept (control group). The operation time, intraoperative blood loss, the time of first anal exhaust and first postoperative off-bed activity, postoperative length of stay, and incidence of postoperative complications were recorded in both groups. The pain of patients was assessed using VAS system. The nausea and vomiting and abdominal distension were assessed using the NVDS and abdominal distension score, respectively, within 24 h after operation. The patient's daily living ability was evaluated by the ADL scale at 3 d after the operation. The time of first anal exhaust, the time of first postoperative off-bed activity time, and the postoperative in-hospital time were all significantly shorter in the ERAS group than those in the control group (P < 0.001). The VAS score in the ERAS group was significantly lower than that in the control group at 12 h, 24 h, 48 h, and 72 h after operation (P < .001). The ERAS group had significantly lower NVDS score and abdominal distension score than the control group (P < 0.001). The postoperative ADL score in the ERAS group was significantly higher than that in the control group (P < 0.001). ERAS during the perioperative period of total laparoscopic radical gastrectomy can promote the postoperative rehabilitation of patients and alleviate postoperative pain and gastrointestinal reactions, which is safe and effective.


Assuntos
Recuperação Pós-Cirúrgica Melhorada , Laparoscopia , Perda Sanguínea Cirúrgica , Gastrectomia , Humanos , Tempo de Internação , Complicações Pós-Operatórias/prevenção & controle
7.
Med Sci Monit ; 25: 10122-10128, 2019 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-31884511

RESUMO

BACKGROUND MicroRNA (miR)-106a was involved in the tumorigenesis and highly expressed in gastric cancer. Required apatinib resistance greatly limits its efficacy in patients. Thus, the aim of the present study was to investigate the potential role of miR-106a-3p in gastric cancer cells with apatinib-resistance. MATERIAL AND METHODS The expression of miR-106a-3p was quantified by real-time quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8 (CCK-8) assay was performed to analyze the sensitivity of gastric cancer cells to apatinib. The expression of relevant drug-resistant proteins was detected by western blot. We searched Targetscan6.2 to find out the target gene of miR-106a-3p. Luciferase reporter assay was used to analyze whether miR-106a-3p bound to relevant gene of SOCS family. The SOCS2, SOCS4, and SOCS5 were qualified by western blot, and their mRNA levels were detected by RT-qPCR. Further, JAK2, STAT3, and their phosphorylation levels were detected by western blot. RESULTS The results showed that the expression of miR-106a-3p was increased in apatinib­resistant gastric cancer, while miR-106a-3p inhibitor reduced the drug-resistance of SGC-7901-AP cells to apatinib. Dual luciferase reporter gene assay suggested that SOCS2, SOCS4, and SOCS5 were target genes of miR-106a-3p. The relevant SOCS genes silencing reversed the effects of miR-106a-3p inhibitor on decreasing the apatinib resistance of SGC-7901-AP cells, while the phosphorylation level of JAK and STAT reduced by miR-106a-3p inhibitor were increased. CONCLUSIONS miR-106a-3p induces apatinib resistance and activates JAK2/STAT3 by targeting SOCS system in gastric cancer. miR-106a-3p/SOCS plays a potent role in gastric cancer cell resistance to apatinib.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Janus Quinase 2/metabolismo , MicroRNAs/metabolismo , Piridinas/farmacologia , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/genética , Fosforilação/efeitos dos fármacos , Neoplasias Gástricas/patologia , Proteínas Supressoras da Sinalização de Citocina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA