Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Adicionar filtros








Intervalo de ano
1.
Chinese Pharmacological Bulletin ; (12): 299-307, 2024.
Artigo em Chinês | WPRIM | ID: wpr-1013590

RESUMO

Aim To explore the new mechanism of triptolide (TRI) inhibiting the progression of hepatocellular carcinoma (HCC) . Methods Different concentrations (0, 0 . 5, 2, and 8 jjunol • L~) of TRI were administered to act on liver cancer cells, and then the cell phenotypes and possible mechanisms were explored using experimental methods such as CCK-8, cell cloning, Transwell, and protein immunoblotting; siRNA was used to interfere with the target gene GSDME and its role was determined. Finally, the mechanism of TRI inhibiting the growth of HCC cells in vivo was validated using a transplanted tumor model. Results TRI could inhibit the proliferation, cloning, and invasion of HCC cells, and promote cell apoptosis. Immunoblotting results showed that the expression of GSDME was significantly upregulated in HepG2 or He-pal-6 hepatocellular carcinoma after TRI treatment, while the expression of cleaved caspase-3 and PARP also significantly increased. Knocking out GSDME could partially reverse TRI-induced cell apoptosis. At the same time, cells knocked down by GSDME had stronger cloning and migration abilities, and the apoptosis rate was reduced compared to the TRI treatment group alone. In vivo experiments showed that TRI inhibited HCC tumor growth, and the TRI + siGSDME group had a faster tumor growth rate than the TRI treatment group alone did. In addition, after TRI stimulation, p-eIF2a and ATF4 in HepG2 and Hepal-6 cells significantly increased. The immunofluorescence results showed a dose-dependent increase in the number of ATF4 positive cells in HepG2 and Hepal-6 cells after TRI stimulation. Conclusion The inhibitory effect of TRI on the growth and invasion of liver cancer cells may be related to its regulation of the ATF4/caspase-3/GSDME signaling pathway and promotion of liver cancer cell apoptosis.

2.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 56-64, 2024.
Artigo em Chinês | WPRIM | ID: wpr-1013340

RESUMO

ObjectiveTo investigate the mechanism of salvianolic acid F (Sal F) in repairing the high glucose-induced injury in human kidney-2 (HK-2) cells via the B-cell lymphoma-2 (Bcl-2)-associated X protein (Bax)/cysteinyl aspartate-specific proteinase 3 (Caspase-3)/gasdermin-E (GSDME) pathway. MethodThe cell counting kit-8 (CCK-8) was used to measure the relative viability of HK-2 cells exposed to high glucose and different concentrations (2.5, 5, 10, 20 μmol·L-1) of Sal F and the relative viability of HK-2 cells treated with Sal F for different time periods. The levels of lactate dehydrogenase (LDH) and interleukin-1β (IL-1β) in the supernatant of the cell culture were measured by the LDH assay kit and enzyme-linked immunosorbent assay (ELISA) kit, respectively. Flow cytometry combined with Annexin V-FITC/propidium iodide (PI) and Hoechst 33342/PI staining was employed to reveal the proportion of PI-positive HK-2 cells exposed to high glucose. Western blotting was employed to determine the protein levels of Bax, Bcl-2, cytochrome C, cysteinyl aspartate-specific proteinase (Caspase)-9, Caspase-3, and GSDME in the HK-2 cells exposed to high glucose and treated with Sal F. The 2,7-dichlorodihydrofluorescein diacetate fluorescence probe (DCFH-DA) and mitochondrial membrane potential assay kit (JC-1) were used to determine the production of reactive oxygen species (ROS) and the mitochondrial membrane potential in the HK-2 cells exposed to high glucose and treated with Sal F. ResultCompared with the blank group, the model group showed decreased cell viability (P<0.01), elevated levels LDH and IL-1β, increased proportion of PI-positive cells (P<0.01), up-regulated protein levels of Bax, cytochrome C, Caspase-9, Caspase-3, and GSDME (P<0.01), down-regulated protein level of Bcl-2 (P<0.01), decreased mitochondrial membrane potential, and excessive ROS accumulation. Compared with the model group, Sal F repaired the high glucose-induced injury in HK-2 cells (P<0.05), lowered the levels of LDH and IL-1β (P<0.05, P<0.01), and decreased the proportion of PI-positive cells (P<0.01). In addition, Sal F down-regulated the protein levels of Bax, cytochrome C, Caspase-9, Caspase-3, and GSDME and up-regulated the protein level of Bcl-2 (P<0.05, P<0.01), increased the mitochondrial membrane potential, and decreased the accumulation of ROS in HK-2 cells. ConclusionSal F can reduce the production of ROS, restore the balance of mitochondrial membrane potential, and inhibit pyroptosis via the Bax/Caspase-3/GSDME signaling pathway to repair the high glucose-induced injury in HK-2 cells.

3.
Journal of Clinical Otorhinolaryngology Head and Neck Surgery ; (12): 30-37, 2024.
Artigo em Chinês | WPRIM | ID: wpr-1011098

RESUMO

Objective:To dentify the genetic and audiological characteristics of families affected by late-onset hearing loss due to GSDMEgene mutations, aiming to explore clinical characteristics and pathogenic mechanisms for providing genetic counseling and intervention guidance. Methods:Six families with late-onset hearing loss from the Chinese Deafness Genome Project were included. Audiological tests, including pure-tone audiometry, acoustic immittance, speech recognition scores, auditory brainstem response, and distortion product otoacoustic emission, were applied to evaluate the hearing levels of patients. Combining with medical history and physical examination to analyze the phenotypic differences between the probands and their family members. Next-generation sequencing was used to identify pathogenic genes in probands, and validations were performed on their relatives by Sanger sequencing. Pathogenicity analysis was performed according to the American College of Medical Genetics and Genomics Guidelines. Meanwhile, the pathogenic mechanisms of GSDME-related hearing loss were explored combining with domestic and international research progress. Results:Among the six families with late-onset hearing loss, a total of 30 individuals performed hearing loss. The onset of hearing loss in these families ranged from 10 to 50 years(mean age: 27.88±9.74 years). In the study, four splicing mutations of the GSDME were identified, including two novel variants: c. 991-7C>G and c. 1183+1G>T. Significantly, the c. 991-7C>G was a de novo variant. The others were previously reported variants: c. 991-1G>C and c. 991-15_991-13del, the latter was identified in three families. Genotype-phenotype correlation analysis revealed that probands with the c. 991-7C>G and c. 1183+1G>T performed a predominantly high-frequency hearing loss. The three families carrying the same mutation exhibited varying degrees of hearing loss, with an annual rate of hearing deterioration exceeding 0.94 dB HL/year. Furthermore, follow-up of interventions showed that four of six probands received intervention(66.67%), but the results of intervention varied. Conclusion:The study analyzed six families with late-onset non-syndromic hearing loss linked to GSDME mutations, identifying four splicing variants. Notably, c. 991-7C>G is the first reported de novo variant of GSDME globally. Audiological analysis revealed that the age of onset generally exceeded 10 years,with variable effectiveness of interventions.


Assuntos
Humanos , Adolescente , Adulto Jovem , Adulto , Criança , Perda Auditiva Neurossensorial/diagnóstico , Surdez/genética , Mutação , Perda Auditiva/genética , Linhagem
4.
Journal of International Oncology ; (12): 285-289, 2023.
Artigo em Chinês | WPRIM | ID: wpr-989559

RESUMO

Pyroptosis is a type of programmed cell death distincted from apoptosis and necrosis, which is accompanied by the lysis of cell membranes and the release of cell contents. Pyroptosis occurs as mediated by Gasdermin protein family and is dependent on the activity of caspase. GSDME is one of the most important members of the Gasdermin protein superfamily. GSDME-mediated pyroptosis relies on the activity of caspase-3. In recent years, with further research on pyroptosis, the mechanism of GSDME-induced pyroptosis is becoming clear. Numerous studies have shown that GSDME-mediated pyroptosis plays an important role in the occurrence and development of tumors, as well as chemotherapy resistance. However, GSDME-mediated pyroptosis has no specificity and can induce pyroptosis of normal cells in the body while inducing tumor cell pyroptosis, thus causing different degrees of damage to various organs of the body. Further study on the mechanism of GSDME-induced pyroptosis, the role of GSDME in malignant tumors and the adverse reactions of chemotherapy can provide new ideas for tumor monitoring, treatment and prognosis judgment.

5.
China Journal of Chinese Materia Medica ; (24): 3589-3601, 2023.
Artigo em Chinês | WPRIM | ID: wpr-981490

RESUMO

This study aimed to explore the anti-glioma effect of natural compound pterostilbene(PTE) through regulating pyroptosis and apoptosis pathways, and to analyze the possible anti-glioma pathways and targets of PTE by network pharmacology and molecular docking. In this study, the action targets of PTE and the glioma targets were obtained by network pharmacology to construct a target network and a protein-protein interaction(PPI) network to predict the possible action targets of PTE against glioma. Molecular docking was performed on the core targets by AutoDock and the action pathways of PTE against glioma were predicted by enrichment analysis. In addition, the effect of PTE on the viability of U87MG and GL261 glioma cells was detected by CCK-8 assay. Clone formation assay and cell scratching assay were used to explore the effect of different concentrations of PTE on the proliferation and migration, respectively of glioma cells. Hoechst staining was used to observe PTE-induced apoptosis in glioma cells. The changes in mitochondrial membrane potential were detected by JC-1 staining. The pyroptosis-inducing effect of PTE on glioma cells was observed by inverted microscopy and lactate dehydrogenase(LDH) assay. Hoechst 33342/PI dual staining assay was performed to detect the integrity of glioma cell membranes. The expressions of pyroptosis and apoptosis-related proteins in glioma cells after PTE induction were determined by Western blot. In this study, 37 anti-glioma targets of PTE were obtained, and enrichment analysis suggested that PTE exerted anti-glioma effects through various signaling pathways including cancer pathway, proteoglycan in cancer, PI3K/AKT pathway, and apoptosis regulatory pathway. Molecular docking revealed that PTE had good binding activity with the main targets. Compared with the control group, PTE significantly reduced the viability as well as the proliferation, migration and adhesion abilities of U87MG and GL261 cells; it induced the apoptosis of the two glioma cells and the decrease of mitochondrial membrane potential in U87MG cells, and the effects increased with the increase of drug concentration. Compared with the conditions in the control group, glioma cells in the PTE group had increased pyroptosis-specific appearance and gradually increased LDH release; the number of PI positive cells was significantly elevated with the increase of PTE concentration as revealed by Hoechst 33342/PI staining; the expression levels of apoptosis-related factors cleaved PARP1 and B-cell lymphoma-2(Bcl-2) associated X(BAX) in the PTE group were markedly up-regulated, while the expression level of Bcl-2 was markedly down-regulated; the activation levels of pyroptosis-related proteins cleaved caspase-3 and gasdermin E-N(GSDME-N) had a remarkable rise in the PTE group, while no significant changes were found in the activation levels of gasdermin D-N(GSDMD-N) and cleaved caspase-1. In summary, PTE plays an anti-glioma role by inhibiting cell viability, proliferation, and migration and activating the caspase-3/GSDME-mediated pyroptosis pathway and mitochondrial apoptosis pathway.


Assuntos
Piroptose , Caspase 3/metabolismo , Farmacologia em Rede , Gasderminas , Simulação de Acoplamento Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Apoptose , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
6.
Chinese Journal of Cancer Biotherapy ; (6): 489-496, 2023.
Artigo em Chinês | WPRIM | ID: wpr-977724

RESUMO

@#[摘 要] 目的:探究细胞焦亡相关基因GSDME在鼻咽癌组织中的表达及其对肿瘤微环境(TME)和患者预后的影响。方法:从TCGA数据库获得548例鼻咽癌患者的基因表达数据及临床资料,运用R语言对GSDME进行差异表达分析、GO-KEGG富集分析;从人类蛋白质图谱(HPA)数据库中获取GSDME蛋白在鼻咽癌组织和相应癌旁组织中的表达数据,通过STRING数据库探究与GSDME相互作用的蛋白质网络;应用ssGSEA算法分析GSDME在鼻咽癌组织中的表达及其与24种免疫细胞浸润的相关性,运用Spearman法分析GSDME与免疫检查点分子的相关性,通过TISIDB数据库分析GSDME表达与细胞因子基因表达的相关性。单因素和多因素COX回归分析筛选预后风险因素,基于风险因素GSDME绘制预后预测列线图和校准图,根据GSDME的表达水平对鼻咽癌患者进行生存分析和风险分析。qPCR法验证中国人鼻咽癌组织中GSDME及4种趋化因子mRNA的表达。结果:数据库数据分析显示,与癌旁组织相比,鼻咽癌组织中GSDME呈高表达(P<0.01),GO-KEGG富集分析显示GSDME参与免疫反应、细胞焦亡相关信号通路,GSDME表达与免疫细胞浸润、细胞因子及免疫检查点分子表达等相关,qPCR检查结果验证了中国人鼻咽癌组织中GSDME呈高表达。GSDME表达量、N分期和M分期是鼻咽癌患者预后的风险因素,基于风险因素建立的列线图和校正图能较好地预测鼻咽癌患者OS,鼻咽癌组织中GSDME高表达的患者预后差。结论:GSDME在鼻咽癌癌组织中呈高表达且是患者预后风险因素,基于GSDME建立的列线图预测预后效能良好;GSDME高表达与TME免疫浸润有关,其可能是免疫治疗的潜在靶点。

7.
Chinese Journal of Cancer Biotherapy ; (6): 318-323, 2023.
Artigo em Chinês | WPRIM | ID: wpr-972720

RESUMO

@#[摘 要] 目的:探讨新补骨脂异黄酮(NBIF)对肝细胞癌(HCC)Huh-7细胞焦亡的影响及其分子机制。方法:体外培养Huh-7细胞,用CCK-8法检测不同浓度的NBIF处理48 h时对细胞存活率的影响,光学显微镜下观察NBIF处理后Huh-7细胞的形态变化,乳酸脱氢酶(LDH)释放实验检测细胞的LDH释放量,WB实验检测细胞中GSDME、caspase-3的蛋白水平变化。采用siRNA干扰Huh-7细胞中caspase-3、GSDME表达后,CCK-8法检测NBIF处理对细胞存活率的影响,WB实验检测GSDME蛋白表达水平,观察NBIF处理对细胞形态的影响,并检测细胞LDH释放量。结果:60 μmol/L以上的NBIF均能显著抑制Huh-7细胞的增殖(均P<0.01),光学显微镜下观察到NBIF处理后的细胞出现肿胀、吐泡现象,且LDH释放增加(P<0.01);WB实验结果表明,NBIF能够激活caspase-3蛋白并切割GSDME蛋白,增加GSDME-N的表达(均P<0.01)。干扰caspase-3、GSDME表达后,NBIF对细胞的抑制作用减弱(均P<0.01),GSDME-N蛋白表达受到抑制(P<0.01),显微镜下细胞肿胀、吐泡现象几乎消失,LDH释放明显减少(P<0.05)。结论:NBIF能够通过caspase-3/GSDME途径诱导Huh-7细胞发生焦亡,从而抑制HCC细胞的增殖,为HCC的治疗提供一种新思路。

8.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 212-220, 2023.
Artigo em Chinês | WPRIM | ID: wpr-976556

RESUMO

Pyroptosis, a new type of inflammatory programmed cell death, is different from apoptosis, necrosis, cytosis, ferroptosis, and autophagy. Pyroptosis is dependent on the activation of cysteine aspartate-specific protease (Caspase), which cleaves key mediator proteins to form pores in the cell membrane and induces the maturation and release of the proinflammatory cytokines interleukin-1β and interleukin-18 into the extracellular environment, resulting in a cascade of inflammatory reactions. Gastric cancer as a malignant tumor of the digestive tract is refractory and has poor prognosis, and the chemoradiotherapy of this disease may lead to a variety of complications. At present, the pathogenesis of gastric cancer remains unclear. Studies have proved that pyroptosis is associated with the occurrence and development of gastric cancer, which has attracted wide attention. Pyroptosis is a double-edged sword for gastric cancer. On the one hand, it can release the contents of proinflammatory cells to amplify or maintain inflammation and induce the "inflammation-cancer" transformation of cells. On the other hand, pyroptosis can enhance the sensitivity of drugs for chemotherapy to improve the therapeutic effect and survival. In recent years, the anti-tumor mechanism of traditional Chinese medicine (TCM) has become a research hotspot as TCM has demonstrated significant effects in clinical application. Therefore, the regulation of pyroptosis by TCM may be a new direction for the treatment of gastric cancer in the future. Based on the available studies, this paper introduces the roles of pyroptosis-associated key proteins in the occurrence and development of gastric cancer. Furthermore, this paper summarizes the effects of TCM prescriptions and active ingredients on alleviating gastric mucosal damage, reducing the incidence of gastric cancer, and preventing tumor metastasis and recurrence by mediating pyroptosis pathways, aiming to provide new ideas for deciphering the mechanism of pyroptosis and exploring the TCM treatment of gastric cancer in the future.

9.
Chinese Journal of Experimental Traditional Medical Formulae ; (24): 226-238, 2023.
Artigo em Chinês | WPRIM | ID: wpr-975176

RESUMO

Pyroptosis, an atypical new cell death mode other than apoptosis and necrosis, has been discovered in recent years. Pyroptosis depends on the cleavage of gasdermins (GSDMs) by Caspases. The activated GSDMs act on the plasma membrane to form a perforation, which results in cell lysis and triggers inflammation and immune response. Pyroptosis can be induced by four distinct signaling pathways, including canonical and non-canonical inflammasome pathways, apoptosis-associated Caspases-mediated pathway, and granzyme pathway. In these signaling pathways, GSDMs are the executors of pyroptosis. Pyroptosis is associated with the death of tumor cells and the inflammatory damage of normal tissues. Recent studies have demonstrated that moderate pyroptosis can lead to tumor cell death to exert an anti-tumor effect, and meanwhile stimulate the tumor immune microenvironment, while it can promote tumor development. Despite the good performance, drug-based anti-tumor therapies such as tumor immunotherapy, chemotherapy, and targeted therapy have some shortcomings such as drug resistance, recurrence, and damage to normal tissues. The latest research shows that a variety of natural compounds have anti-tumor effects in the auxiliary treatment of tumors by mediating the pyroptosis pathways in a multi-target and multi-pathway manner, which provide new ideas for the study of anti-tumor therapy. We reviewed the molecular mechanism of pyroptosis and the regulatory role of pyroptosis in tumors and tumor immune microenvironment, and summarized the recent research progress in the natural medicinal components regulating pyroptosis in anti-tumor therapy, with a view to providing ideas for the research on the anti-tumor therapy based on pyroptosis.

10.
Chinese Journal of Cancer Biotherapy ; (6): 251-257, 2022.
Artigo em Chinês | WPRIM | ID: wpr-923464

RESUMO

@#[摘 要] 细胞焦亡是近年来发现的一种新型细胞死亡的方式,是一种受焦孔素(GSDM)家族调控的炎症性程序性细胞死亡,其主要特征是膜穿孔、细胞肿胀及细胞破裂。细胞焦亡发生的机制分为由GSDMD介导的Caspase-1和Caspase-4/-5/-11依赖性经典炎症小体途径和由GSDME介导的Caspase-3和颗粒酶依赖性非经典炎症小体途径等。近年来研究显示,细胞焦亡具有抑制和促进肿瘤发生发展的双重作用,并且细胞焦亡的诱导在抗肿瘤免疫治疗中也发挥双重效应:一方面通过促进炎症因子释放,形成肿瘤微环境,抑制抗肿瘤免疫,另一方面则通过引发抗肿瘤炎症反应抑制肿瘤细胞的增殖。此外,细胞焦亡的诱导在化疗及其他联合治疗中也发挥着重要作用。进一步研究发现,中药及其提取物调控细胞焦亡的诱导对于治疗肿瘤至关重要。

11.
Chinese Journal of Endocrine Surgery ; (6): 150-154, 2022.
Artigo em Chinês | WPRIM | ID: wpr-930317

RESUMO

Objective:To explore the mechanism by which GSDME promoter methylation regulates the chemotherapy sensitivity of breast cancer (BC) cells through pyrolysis.Methods:Tissues of 54 cases with BC treated with chemotherapy were collected and divided into chemotherapy resistant group and sensitive group. Docetaxel (DTX) was used to treat BC cells to establish drug-resistant BC cells. CCK8 was used to detect cell resistance.qRT-PCR was used to detect the expression of GSDME in tissues and cells. Then sulfite sequencing method was utilized to determine the methylation level of the GSDME promoter methylation site. The expression level of GSDME in DTX-treated BC cells was intervened, and qRT-PCR was used to detect the expression level of Caspase-1, Caspase-4, IL-1β, and IL-18 in cells to assess the degree of cell pyrolysis.Results:Compared with the chemotherapy sensitive group, the expression of GSDME was down-regulated in the chemotherapy resistant group ( t=6.54, P<0.001) . Compared with MCF-10A cells, Caspase-1 ( t=9.14, P<0.001) , Caspase-4 ( t=9.35, P<0.001) , IL-1β ( t=6.13, P=0.004) , and IL-18 ( t=5.49, P=0.005) expression level in MCF-10A/DTX cells were decreased, however, the above indicators were up-regulated after overexpression of GSDME in MCF-10A/DTX cells. Hypermethylation in GSDME promoter region led to a decrease in mRNA expression ( t=12.56, P<0.001) . In tolerance tissues, the methylation level in the GSDME promoter region was negatively correlated with the mRNA level ( r=-0.57, P=0.010) . After treating MCF-10A/DTX cells with 5-azacytidine, a methylation inhibitor,Caspase-1, Caspase-4, IL-1β, IL-18 levels were significantly increased, and GSDME level were up-regulated. Conclusion:Hypermethylation in the promoter region of GSDME leads to decreased mRNA expression, inhibits pyrolysis of BC cells, and reduces the sensitivity of DTX chemotherapy.

12.
Chinese Journal of Cancer Biotherapy ; (6): 288-293, 2021.
Artigo em Chinês | WPRIM | ID: wpr-876058

RESUMO

@#[摘 要] 细胞焦亡(pyroptosis)是新近发现的一种依赖炎性半胱天冬酶(caspase)的促炎程序性细胞死亡方式(regulated cell death,RCD),主要通过gasdermin家族成员发生剪切活化,引起细胞膜穿孔和细胞内容物释放的过程。Gasdermin E(GSDME)是gasdermin家族的主要成员之一,近来研究发现在几种常见的肿瘤中常因其启动子甲基化而低表达,进而增强了肿瘤的增殖和转移能力;分析其结构和功能显示,GSDME可被激活态的caspase-3切割并形成具有造孔活性的GSDME-N末端结构域,从而诱导肿瘤细胞发生焦亡,介导肿瘤细胞死亡。本文就GSDME介导焦亡的机制及其与肿瘤发生发展的关系作一综述,以期提出新的临床肿瘤诊疗方向。

13.
Chinese Journal of Cancer Biotherapy ; (6): 884-888, 2020.
Artigo em Chinês | WPRIM | ID: wpr-825119

RESUMO

@#[Abstract] Objective: To observe the pyrolysis of colorectal cancer Lovo cells overexpressing Gasdermin E (GSDME) after the treatment with oxaliplatin. Methods: The expression level of GSDME gene in colorectal cancer Lovo cells and normal colorectal epithelial HCOEPIC cells was detected by qPCR. The GSDME-WT (wild-type GSDME) and GSDME-D270A (mutant GSDME) recombinant plasmids were constructed. The plasmids were packaged as lentivirus and then transfected into Lovo cells to construct Lovo cell line with stable and high expression of GSDME. Western blotting was used to detect the expression level of GSDME in cells of WT, D270A and empty vector groups. Different concentrations of oxaliplatin (0, 4, 8, 16, 32, 64 µg/ml) were applied to treat Lovo cells and HCOEPIC cells in WT and D270A groups, and the morphological changes of the cells were observed under a microscope. Results: The expression of GSDME in HCOEPIC cells was significantly higher than that in Lovo cells (P<0.01). GSDME-WT and GSDME-D270A plasmids with high GSDME expression and the corresponding Lovo cell lines were successfully constructed. Compared with the empty vector group, the expression level of GSDME in Lovo cells of WT and D270A groups were significantly increased (all P<0.05). Observation under the microscope showed that after being treated with 64 µg/ml oxaliplatin for 9 and 12 hours, the volume of Lovo cells and HCOEPIC cells in WT group gradually increased and “blistered” to one side and showed obvious pyrolysis phenomenon. The pyrolysis rate of cells in WT group was significantly higher than that of the control group without oxaliplatin treatment (Lovo cells: [7.405±1.010]% vs [3.441±0.401]%, P<0.05; HCOEPIC cells: [7.203±1.020]% vs [4.201±0.302]%, P<0.05). Conclusion: Oxaliplatin promotes the pyrolysis of colorectal cancer Lovo cells overexpressing GSDME gene.

14.
Chinese Journal of Cancer Biotherapy ; (6): 146-151, 2019.
Artigo em Chinês | WPRIM | ID: wpr-792957

RESUMO

@# Objective: To investigate whether GSDME affects the sensitivity of breast cancer MCF-7 cells to paclitaxel (PTX) by regulating cell pyroptosis. Methods: GSDME was knocked-down in MCF-7 cells by RNA interference technique. CCK-8 assay, flow cytometry,lactate dehydrogenase (LDH) release method and Wb were respectively used to detect cell proliferation, pyroptotic rate, LDH release, GSDME-N-terminal protein and cleaved-caspase-3 protein levels in PTX-treated MCF-7 cells before and after GSDME knockdown. Results: Compared with the control group, the pyroptotic rate, LDH release, GSDME-N-terminal protein and cleaved-caspase-3 protein levels in the PTX-treatment group significantly increased (all P<0.01). Compared with the si-NC group, the PTX-sensitivity of si-GSDME group decreased, and the pyroptotic rate, LDH release and GSDME-N-terminal protein all significantly decreased (all P< 0.01). Conclusion: Knock-down of GSDME in MCF-7 cells significantly inhibited cell pyroptosis and reduced drug sensitivity of MCF7 cells to PTX.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA