Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Ther ; 18(5): 1380-1386, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36204886

RESUMO

Objective: Lung cancer is currently the cancer with the highest incidence and death toll worldwide. Hydrogen gas has been found to affect a variety of diseases; however, the effect of hydrogen gas on patients with lung cancer has not been reported. Therefore, we determined the effect of hydrogen gas on apoptosis of lung adenocarcinoma in vivo and in vitro. Materials and Methods: A549 cells in the logarithmic phase were treated with 20%, 40%, or 60% hydrogen gas. Cell apoptosis was evaluated by flow cytometry. The A549 cell suspension was inoculated into 15 nude mice. The mice were randomly divided into control, hydrogenation (inhalation of 60% hydrogen gas), and cisplatin groups (intraperitoneal injection of cisplatin [4 mg/kg]). After 3 weeks, the tumor tissue was removed and measured. We identified differentially expressed genes by transcriptional profiling. The levels of X-linked inhibitor of apoptosis (XIAP), baculoviral inhibitor of apoptosis protein repeat-containing 3 (BIRC3), and BCL2-associated X and apoptosis regulator (BAX) protein expression were detected by Western blotting and immunohistochemistry. Results: Compared with the control group, the apoptosis rates in the 20%, 40%, and 60% hydrogen gas groups were significantly increased (P < 0.01). The levels of XIAP and BIRC3 protein expression were clearly decreased in the hydrogen gas group compared to the control group. Moreover, cisplatin and hydrogen gas reduced the tumor volume in nude mice (P < 0.01). Transcriptome sequencing showed that XIAP, BIRC2, BIRC3, BAX, PIK3CD, and ATM were related to apoptosis. Hydrogen gas further decreased the levels of XIAP and BIRC3 expression than in nude mice (P < 0.01). Conclusion: Hydrogen gas promoted apoptosis of A549 cells by reducing the expression of XIAP and BIRC3 protein.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Células A549 , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Animais , Apoptose/genética , Proteína 3 com Repetições IAP de Baculovírus , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino , Humanos , Hidrogênio/farmacologia , Proteínas Inibidoras de Apoptose/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos , Camundongos Nus , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteína X Associada a bcl-2/genética
2.
Oncol Lett ; 20(4): 112, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32863925

RESUMO

Our previous study found that hydrogen gas (H2) could efficiently inhibit lung cancer progression; however, the underlying mechanisms still remains to be elucidated. The present study aimed to explore the roles of H2 in lung cancer cell autophagy, and reveal the effects of autophagy on H2-mediated lung cancer cell apoptosis and the underlying mechanisms. The expression levels of proteins associated with cell apoptosis and autophagy were detected using western blot analysis. Cell autophagy was inhibited by 3-methyladenine treatment or Beclin1 downregulation, while rapamycin was used to induce autophagy. Cell growth and apoptosis were detected using the Cell Counting Kit-8 and flow cytometry assays, respectively. The results demonstrated that cell apoptosis and autophagy were significantly enhanced in the A549 and H1975 lung cancer cell lines treated with H2. However, autophagy enhancement weakened H2 roles in promoting cell apoptosis and vice versa. In addition, it was found that H2 treatment induced marked decreases in the protein expression levels of phosphorylated STAT3 and Bcl2, and overexpression of STAT3 abolished H2 roles in promoting cell apoptosis and autophagy. Overall, the present study revealed that H2 can promote lung cancer cell apoptosis and autophagy via inhibiting the activation of STAT3/Bcl2 signaling and suppression of autophagy can enhance H2 roles in promoting lung cancer cell apoptosis.

3.
Biosci Rep ; 40(4)2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32314789

RESUMO

Hydrogen gas (H2) has been identified to play an anti-tumor role in several kinds of cancers, but the molecular mechanisms remain largely unknown. In our previous study, our project group found that H2 could decrease the expression of CD47 in lung cancer A549 cells via the next-generation sequencing, indicating that CD47 might be involved in H2-mediated lung cancer repression. Therefore, the present study aimed to explore the effects of CD47 on H2-induced lung cancer repression. Western blotting and real-time PCR (RT-PCR) assays were used to detect the levels of proteins and mRNAs, respectively. Cell proliferation, invasion, migration and apoptosis were detected by using the cell counting kit-8 (CCK-8), Transwell chambers, wound healing and flow cytometry assays, respectively. The results showed that H2 treatment caused decreases in the expression levels of CD47 and cell division control protein 42 (CDC42) in a dose-dependent manner. Up-regulation of CD47 abolished H2 roles in promoting lung cancer cell apoptosis and repressing cell growth, invasion and migration in both A549 and H1975 cell lines. However, knockdown of CD47 enhanced H2 role in lung cancer inhibition. Moreover, we also observed that H2 treatment induced obvious inhibitions in the expression levels of CDC42 and CD47 in mice tumor tissues, as well as reinforced macrophage-mediated phagocytosis in A549 and H1975 cells. In conclusion, the current study reveals that H2 inhibits the progression of lung cancer via down-regulating CD47, which might be a potent method for lung cancer treatment.


Assuntos
Antígeno CD47/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hidrogênio/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Células A549 , Administração por Inalação , Animais , Apoptose/efeitos dos fármacos , Antígeno CD47/genética , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Fagocitose/efeitos dos fármacos , Fagocitose/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína cdc42 de Ligação ao GTP/metabolismo
4.
Biomed Pharmacother ; 111: 588-595, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30605882

RESUMO

PYCR1 is over-expressed in non-small-cell lung cancer (NSCLC) and its high expression accelerates the progression of NSCLC. However, the underlying mechanisms of PYCR1 in NSCLC progression remain poorly understood. Our study determined the mechanisms of PYCR1 in promotion of the occurrence and development of NSCLC in vitro and in vivo. Firstly, the expression patterns of PYCR1 in NSCLC tissues and cells were determined by RT-PCR, western blot and immunohistochemistry. Then, the effects of PYCR1 on cell proliferation and apoptosis were evaluated by CCK-8 and flow cytomery assays. Finally, we explored the up-regulatory microRNAs (miRs) of PYCR1 and determined if MAPK pathway involved in this process. PYCR1 expression was elevated in NSCLC tissue samples and cells, and the high expression of PYCR1 closely associated with patients' advanced clinical process and poor outcome. Up-regulation of PYCR1 significantly increased the expression of p38 and promoted its nuclear accumulation. Besides, PYCR1 expression was negatively regulated by miR-488, and up-regulation of miR-488 significantly inhibited cell proliferation and tumorigenesis and increased cell apoptosis, and decreased p38 expression and its nuclear accumulation, whereas up-regulation of PYCR1 rescued these results induced by miR-488 over-expression. Collectively, these data suggest the mechanism of PYCR1 in promotion of NSCLC progression. PYCR1 is negatively regulated by miR-488 and then promotes the occurrence and development of NSCLC and activates p38 MAPK pathway.


Assuntos
Carcinogênese/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Progressão da Doença , Neoplasias Pulmonares/metabolismo , MicroRNAs/biossíntese , Pirrolina Carboxilato Redutases/biossíntese , Células A549 , Idoso , Animais , Carcinogênese/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Pirrolina Carboxilato Redutases/genética , Distribuição Aleatória , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , delta-1-Pirrolina-5-Carboxilato Redutase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA