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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Sleep Breath ; 27(1): 67-75, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35347658

RESUMO

PURPOSE: An earlier study found that heated humidification reduced the side effects of positive airway pressure (PAP) in patients with obstructive sleep apnoea (OSA). However, other studies disagreed with this finding. Therefore, we evaluated the relationship between heated humidification and the side effects of PAP in patients with OSA. METHODS: According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, six databases were searched for relevant randomised controlled trials (RCTs) performed from January 1999 to June 2021. RESULTS: From 1012 retrieved articles, we identified 9 eligible RCTs. Compared to the control group, the heated humidification group reported improvements in dry nose (pooled standardised mean difference [SMD] = - 0.70, 95% confidence interval [CI] = - 0.96 to - 0.45, I2 = 0%, p < 0.00001), blocked nose (SMD = - 0.47, 95%CI - 0.69 to - 0.25, I2 = 36%, p < 0.0001), runny nose (SMD = - 0.22, 95%CI - 0.44 to 0, I2 = 0%, p = 0.05), dry mouth (SMD = - 0.62, 95%CI - 0.87 to - 0.37, I2 = 0%, p < 0.00001), and dry throat (SMD = - 0.61, 95%CI - 0.90 to - 0.33, I2 = 41%, p < 0.0001), but did not enhance positive airway pressure adherence (pooled mean difference [MD] = 0.24, 95% CI - 0.10 to 0.58, I2 = 0%, p = 0.17). CONCLUSIONS: Heated humidification improved some PAP side effects in patients with OSA but did not increase PAP adherence. Additional large-scale multi-centre RCTs are needed to verify the long-term effects.


Assuntos
Obstrução Nasal , Apneia Obstrutiva do Sono , Humanos , Apneia Obstrutiva do Sono/terapia
2.
J Biomed Sci ; 29(1): 34, 2022 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-35655269

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) accounts for the majority of liver cancer cases, while metastasis is considered the leading cause of HCC-related death. However, the currently available treatment strategies for efficient suppression of metastasis are limited. Therefore, novel therapeutic targets to inhibit metastasis and effectively treat HCC are urgently required. METHODS: Wound healing and Transwell assays were used to determine the migration and invasion abilities of HCC cells in vitro. Quantitative real-time PCR (qRT-PCR), protein array, immunofluorescence, and immunoprecipitation experiments were used to study the mechanism of DYRK1A-mediated metastasis. A tail vein metastasis model and H&E staining were utilized to assess metastatic potential in vivo. RESULTS: The results of the current study demonstrated that dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) was upregulated in HCC tissues compared with normal liver tissues. Additionally, the level of DYRK1A was increased in primary HCC tissues of patients with metastasis compared with those of patients without metastasis, and DYRK1A overexpression correlated with worse outcomes in liver cancer patients. Gain- and loss-of-function studies suggested that DYRK1A enhanced the invasion and migration abilities of HCC cells by promoting epithelial-mesenchymal transition (EMT). Regarding the promoting effect of DYRK1A on cell invasion, the results showed that DYRK1A was coexpressed with TGF-ß/SMAD and STAT3 signalling components in clinical tumour samples obtained from patients with HCC. DYRK1A also activated TGF-ß/SMAD signalling by interacting with tuberous sclerosis 1 (TSC1) and enhanced metastasis of HCC cells by activating STAT3. Furthermore, DYRK1A promoted EMT by cooperatively activating STAT3/SMAD signalling. CONCLUSION: Overall, the present study not only uncovered the promoting effect of DYRK1A on HCC metastasis and revealed the mechanism but also provided a new approach to predict and treat metastatic HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Humanos , Neoplasias Hepáticas/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
3.
Anticancer Drugs ; 32(7): 727-733, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33735117

RESUMO

Vinpocetine is widely used to treat cerebrovascular diseases. However, the effect of vinpocetine to treat hepatocellular carcinoma (HCC) has not been investigated. In this study, we revealed that vinpocetine was associated with antiproliferative activity in HCC cells, but induced cytoprotective autophagy, which restricted its antitumor activity. Autophagy inhibitors improved the antiproliferative activity of vinpocetine in HCC cells. Sorafenib is effective to treat advanced HCC, but the effect of autophagy induced by sorafenib is indistinct. We demonstrated vinpocetine plus sorafenib suppressed the cytoprotective autophagy activated by vinpocetine in HCC cells and significantly induced apoptosis and suppressed cell proliferation in HCC cells. In addition, vinpocetine plus sorafenib activates glycogen synthase kinase 3ß (GSK-3ß) and subsequently inhibits cytoprotective autophagy induced by vinpocetine in HCC cells. Meanwhile, overexpression of GSK-3ß was efficient to increase the apoptosis induced by vinpocetine plus sorafenib in HCC cells. Our study revealed that vinpocetine plus sorafenib could suppress the cytoprotective autophagy induced by vinpocetine and subsequently show synergistically anti-HCC activity via activating GSK-3ß and the combination of vinpocetine and sorafenib might reverse sorafenib resistance via the PI3K/protein kinase B/GSK-3ß signaling axis. Thus, vinpocetine may be a potential candidate for sorafenib sensitization and HCC treatment, and our results may help to elucidate more effective therapeutic options for HCC patients with sorafenib resistance.


Assuntos
Glicogênio Sintase Quinase 3 beta/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Sorafenibe/farmacologia , Alcaloides de Vinca/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimioterapia Combinada , Células Hep G2 , Humanos , Transdução de Sinais/efeitos dos fármacos , Sorafenibe/administração & dosagem , Alcaloides de Vinca/administração & dosagem
4.
Sleep Breath ; 25(4): 1761-1771, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33694034

RESUMO

PURPOSE: This meta-analysis was conducted to assess the effects of telemedicine interventions on continuous positive airway pressure (CPAP) adherence in patients with obstructive sleep apnoea (OSA). METHODS: The PubMed, Cochrane Library, OVID, Web of Science and EBSCO host databases were searched from January 2004 to February 2020 for randomised controlled trials (RCTs) that assessed the effects of telemedicine interventions on CPAP adherence in patients with OSA. The study inclusion criteria were RCTs that compared patients who received telemedicine interventions with a control group and reported a change in CPAP adherence. The primary outcome was the improvement in CPAP adherence. RESULTS: In total, there were 11 RCTs (n = 1358) with quantitative analyses. Intervention times ranged from 1 to 6 months. Compared to controls, the telemedicine group exhibited better adherence to CPAP therapy (pooled mean difference (MD) = 0.57, 95% CI = 0.33 to 0.80, I2 = 7%, p < 0.00001). We performed sensitivity analyses by the type of telemedicine intervention, comorbidity burden, and OSA severity to explore whether or not their effect sizes may have affected the time of CPAP application. We performed subgroup analyses by follow-up duration, age, and OSA Epworth sleepiness scale (ESS) symptoms to determine if their effect sizes may have affected the time of CPAP application. However, these analyses did not change the statistical significance of the pooled estimate. CONCLUSIONS: The use of telemedicine for up to 6 months may enhance CPAP adherence in patients with OSA, when compared to no intervention. Our study was searched from January 2004 to February 2020 for randomised controlled trials (RCTs) that assessed the effects of telemedicine interventions on CPAP adherence in patients with OSA. Future studies can continue to search for articles after February 2020.


Assuntos
Pressão Positiva Contínua nas Vias Aéreas , Avaliação de Resultados em Cuidados de Saúde , Cooperação do Paciente , Ensaios Clínicos Controlados Aleatórios como Assunto , Apneia Obstrutiva do Sono/terapia , Telemedicina , Humanos
5.
Biochem Biophys Res Commun ; 512(4): 852-858, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-30929918

RESUMO

Regorafenib has been approved to treat patients who have HCC progression after sorafenib failure, however, regorafenib also faces the risk of drug resistance and subsequent progression of HCC patients. As LSD1 inhibitors can alleviate acquired resistance to sorafenib, in this context, we are interested to investigate the role of LSD1 in regorafenib treatment. Firstly, over-expressed LSD1 was observed in HCC patients and predicted poor prognosis. However, regorafenib failed to suppress the expression of LSD1 in HCC cells. Thus, we hypothesized that LSD1 inhibition could enhance the anti-HCC activity of regorafenib. As expected, LSD1 knockdown could enhance anti-proliferation effect of regorafenib in HCC cells. LSD1 inhibitor SP2509 could enhance the cytotoxic and apoptotic effects of regorafenib in HCC cells. In addition, clinically used LSD1 inhibitor tranylcypromine also enhanced anti-HCC effect of regorafenib. Furthermore, LSD1 suppressed by SP2590 or tranylcypromine could alleviate the activated p-AKT (ser473) induced by regorafenib in HCC cells. Thus, inhibiting LSD1 might be an attractive target for regorafenib sensitization and clinical HCC therapy, our findings could help to elucidate more effective therapeutic options for HCC patients.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Histona Desmetilases/genética , Neoplasias Hepáticas/tratamento farmacológico , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/metabolismo , Humanos , Hidrazinas/administração & dosagem , Hidrazinas/farmacologia , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Compostos de Fenilureia/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/administração & dosagem , Serina/metabolismo , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Tranilcipromina/administração & dosagem , Tranilcipromina/farmacologia
6.
Biochem Biophys Res Commun ; 503(2): 1123-1129, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29944884

RESUMO

Lung squamous cell carcinoma (LSCC) is a common type of non-small-cell lung cancer (NSCLC) and lacks effective treatment. Regorafenib, an oral multikinase inhibitor, has demonstrated promising anti-tumor activity in various solid tumors. To study whether regorafenib inhibits LSCC cells, we investigate the compound in several LSCC cell lines and explore the possible mechanism. In this study, we confirmed that regorafenib had anti-proliferation effect on LSCC cell lines by inducing G0/G1 arrest. In addition, glycogen synthase kinase 3ß (GSK3ß) remained at the same level and Ser9 phosphorylation of GSK3ß decreased with increasing incubation time and increasing regorafenib concentration in LSCC cells. GSK3ß inhibition enhanced the anti-tumor activity of regorafenib. Thus, GSK3ß activation restricted the anti-cancer effect of regorafenib on LSCC. In conclusion, regorafenib might be a promising drug for LSCC therapy. GSK3ß might be a potential target to increase the anti-tumor effect of regorafenib in LSCC cells.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Fosforilação/efeitos dos fármacos
7.
Infect Drug Resist ; 17: 319-327, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38293312

RESUMO

Introduction: Carbapenem-Resistant Enterobacteriaceae (CRE) has posed a significant threat to humans.The aim of this study was to investigate the molecular characteristics of blaKPC-producing Escherichia coli in a university-affiliated tertiary hospital. Methods: Polymerase chain reaction (PCR) and BLAST+ software were used to detect the prevalence of blaKPC in E. coli and Klebsiella pneumoniae. Whole-genome sequencing was performed for the blaKPC-harboring clinical E. coli isolates. Antimicrobial resistance genes, MLSTs, KPC-carrying plasmid typing and genetic environment of blaKPC were analyzed. A maximum likelihood core single nucleotide polymorphism (SNP)-based phylogeny tree was constructed to determine the evolutionary relationships within this ST131 collection. Conjugation experiments were performed to determine the mobilization of blaKPC. The minimal inhibitory concentrations of the common antimicrobial agents were determined using the broth microdilution method. Results: The prevalence of blaKPC in 424 clinical E. coli isolates and 1636 E. coli strains from GenBank database were 2.2% (45/2060) whereas the detection rate of blaKPC in K. pneumoniae from the GenBank database was 29.8% (415/1394). The blaKPC-harboring conjugants exhibited resistance to multiple ß-lactams, except for cefepime-zidebactam and ceftazidime-avibactam. All blaKPC-carring E. coli isolates were susceptible to tigecycline and polymyxin B. ST131 was the dominant sequence type of blaKPC-carring E. coli, accounting for 40.0% (18/45). Most of the blaKPC-producing ST131 E. coli (89.5%,17/19) belonged to clade C ST131 lineage. Genetic environment analysis revealed that 57.8% (26/45) of blaKPC gene was linked to Tn4401-associated structure ISKpn6-blaKPC-ISKpn7. IncN was the most common plasmid type in KPC-producing E. coli whereas IncFII was the dominant plasmid type in KPC-producing K. pneumoniae. Conclusion: The detection rate of blaKPC was lower in E. coli compared with K. pneumoniae. The dominant sequence and plasmid types of blaKPC-harboring isolates differed between E. coli and K. pneumoniae. Further studies about the role of the defense system in acquisition of KPC-plasmids in E. coli will be performed to provide new insights into the low prevalence of blaKPC.

8.
Biochem Pharmacol ; 220: 116011, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38154548

RESUMO

Sinularin, a natural product that purified from soft coral, exhibits anti-tumor effects against various human cancers. However, the mechanisms are not well understood. In this study, we demonstrated that Sinularin inhibited the viability of human prostate cancer cells in a dose-dependent manner and displayed significant cytotoxicity only at high concentration against normal prostate epithelial cell RWPE-1. Flow cytometry assay demonstrated that Sinularin induced tumor cell apoptosis. Further investigations revealed that Sinularin exerted anti-tumor activity through intrinsic apoptotic pathway along with up-regulation of pro-apoptotic protein Bax and PUMA, inhibition of anti-apoptotic protein Bcl-2, mitochondrial membrane potential collapses, and release of mitochondrial proteins. Furthermore, we illustrated that Sinularin induced cell apoptosis via up-regulating PUMA through inhibition of FOXO3 degradation by the ubiquitin-proteasome pathway. To explore how Sinularin suppress FOXO3 ubiquitin-proteasome degradation, we tested two important protein kinases AKT and ERK that regulate FOXO3 stabilization. The results revealed that Sinularin stabilized and up-regulated FOXO3 via inhibition of AKT- and ERK1/2-mediated FOXO3 phosphorylation and subsequent ubiquitin-proteasome degradation. Our findings illustrated the potential mechanisms by which Sinularin induced cell apoptosis and Sinularin may be applied as a therapeutic agent for human prostate cancer.


Assuntos
Proteínas Reguladoras de Apoptose , Diterpenos , Compostos Heterocíclicos com 3 Anéis , Neoplasias da Próstata , Humanos , Masculino , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Proteína Forkhead Box O3 , Próstata/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ubiquitinas/metabolismo
9.
Appl Bionics Biomech ; 2022: 9808334, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35600847

RESUMO

Background: Various noninvasive methods of intracranial pressure (ICP) measurement have been proposed. Each has unique advantages and limitations. This study was aimed at investigating the relationships between lateral ventricular asymmetry on admission computed tomography, optic nerve sheath diameter (ONSD), and ICP in traumatic brain injury (TBI) patients. Methods: A prospective observational study was conducted in the patients admitted to our department between October 2018 and October 2020. 20 patients with moderate-severe TBI with a Glasgow Coma Scale of 3-12 were enrolled. Lateral ventricle volume (LVV) value measurements were conducted using ITK-SNAP software. The lateral ventricular volume ratio (LVR) was quantified by dividing the larger LVV by the smaller. Results: ONSD and LVR had a good correlation with ICP. Admission LVR of >1.735 was shown to have a sensitivity of 90.9% and a specificity of 88.9% for prediction of ICP increase (AUC = 0.879; standard error = 0.091; 95% CI = 0.701 to 1.0; significance level p < 0.004). Admission ONSD of >5.55 mm was shown to have a sensitivity of 81.8% and a specificity of 88.9% for prediction of ICP increase (AUC = 0.919; standard error = 0.062; 95% CI = 0.798 to 1.0; significance level p < 0.002). Combining the ONSD and LVR, the sensitivity could be improved to 90.9% in parallel test, and the specificity could be improved to 100% in serial test. Conclusion: ONSD and LVR measurements can diagnose elevated ICP in traumatic brain injury patients. ONSD combining with LVR may further improve the diagnostic evaluation.

10.
Oncogene ; 39(39): 6203-6217, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32826950

RESUMO

Clear cell renal cell carcinoma (ccRCC) is the most common renal cancer and frequently diagnosed at an advanced stage. It is prone to develop unpredictable metastases even with proper treatment. Antiangiogenic therapy is the most effective medical treatment for metastatic ccRCC. Thus, exploration of novel approaches to inhibit angiogenesis and metastasis may potentially lead to a better therapeutic option for ccRCC. Among all the types of cancer, renal cancer samples exhibited the maximum upregulation of ApoC1 as referred to in the Oncomine database. The expression of ApoC1 was increased accompanied by ccRCC progression. A high level of ApoC1 was closely related to poor survival time in ccRCC patients. Furthermore, ApoC1 was over-expressed in the highly invasive ccRCC cells as compared to that in the low-invasive ccRCC cells. Besides, ApoC1 promoted metastasis of ccRCC cells via EMT pathway, whereas depletion of ApoC1 alleviated these effects. ApoC1 as a novel pro-metastatic factor facilitates the activation of STAT3 and enhances the metastasis of ccRCC cells. Meanwhile, ApoC1 in the exosomes were transferred from the ccRCC cells to the vascular endothelial cells and promoted metastasis of the ccRCC cells via activating STAT3. Finally, the metastatic potential of the ccRCC cells driven by ApoC1 was suppressed by DPP-4 inhibition. Our study not only identifies a novel ApoC1-STAT3 pathway in ccRCC metastasis but also provides direction for the exploration of novel strategies to predict and treat metastatic ccRCC in the future.


Assuntos
Apolipoproteína C-I/metabolismo , Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Fator de Transcrição STAT3/metabolismo , Compostos de Anilina/farmacologia , Apolipoproteína C-I/antagonistas & inibidores , Apolipoproteína C-I/biossíntese , Apolipoproteína C-I/genética , Compostos de Benzilideno/farmacologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Metástase Neoplásica , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Fator de Transcrição STAT3/genética , Análise de Sobrevida , Transcrição Gênica , Células Tumorais Cultivadas
11.
Cancer Biol Med ; 17(2): 387-400, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32587776

RESUMO

Objective: Mcl-1 overexpression confers acquired resistance to Bcl-2 inhibitors in non-small cell lung cancer (NSCLC), but no direct Mcl-1 inhibitor is currently available for clinical use. Thus, novel therapeutic strategies are urgently needed to target Mcl-1 and sensitize the anti-NSCLC activity of Bcl-2 inhibitors. Methods: Cell proliferation was measured using sulforhodamine B and colony formation assays, and apoptosis was detected with Annexin V-FITC staining. Gene expression was manipulated using siRNAs and plasmids. Real-time PCR and Western blot were used to measure mRNA and protein levels. Immunoprecipitation and immunofluorescence were used to analyze co-localization of dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) and Mcl-1. Results: Suppression of DYRK1A resulted in reduced Mcl-1 expression in NSCLC cells, whereas overexpression of DYRK1A significantly increased Mcl-1 expression. Suppression of DYRK1A did not alter Mcl-1 mRNA levels, but did result in an accelerated degradation of Mcl-1 protein in NSCLC cells. Furthermore, DYRK1A mediated proteasome-dependent degradation of Mcl-1 in NSCLC cells, and DYRK1A co-localized with Mcl-1 in NSCLC cells and was co-expressed with Mcl-1 in tumor samples from lung cancer patients, suggesting that Mcl-1 may be a novel DYRK1A substrate. We showed that combined therapy with harmine and Bcl-2 antagonists significantly inhibited cell proliferation and induced apoptosis in NSCLC cell lines as well as primary NSCLC cells. Conclusions: Mcl-1 is a novel DYRK1A substrate, and the role of DYRK1A in promoting Mcl-1 stability makes it an attractive target for decreasing Bcl-2 inhibitor resistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Idoso , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , RNA Interferente Pequeno/genética , Quinases Dyrk
12.
Phytomedicine ; 68: 153189, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32070867

RESUMO

BACKGROUND: NSCLC is the major type of lung cancer and the survival rates of NSCLC patients remain low. AZD9291 is a third-generation EGFR-TKI and approved to treat NSCLC patients harboring EGFR T790M mutation and common targetable activating EGFR mutations, but it has a limited effect for wtEGFR NSCLC. PURPOSE: The current study investigated whether shikonin could enhance the antitumor effect of AZD9291 in wtEGFR NSCLC cells. METHODS: SRB and colony formation assay were used to detect the proliferation of NSCLC cells, propidium iodide staining was performed to detect the apoptosis, ROS was analyzed using DCFH-DA staining, and western blot was used to detect the expression of indicated proteins. RESULTS: We demonstrated that shikonin, a natural ROS inducer, could enhance the antitumor effect of AZD9291 in wtEGFR NSCLC cells. In addition, shikonin increased AZD9291-induced apoptosis accompanying with the generation of ROS and activation of ER stress. Furthermore, ROS inhibition by NAC or GSH reversed the apoptosis induced by shikonin plus AZD9291, and recovered the ER stress activated by combination treatment, indicating that ROS mediated ER stress played a vital role in this combination therapy. Moreover, shikonin increased the anticancer activity of AZD9291 in primary wtEGFR NSCLC cells through ROS-mediated ER stress. CONCLUSION: Our study suggests that combining shikonin with AZD9291 is a promising therapeutic strategy for treating wtEGFR NSCLC patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Células A549 , Acrilamidas/administração & dosagem , Compostos de Anilina/administração & dosagem , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inibidores , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Naftoquinonas/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Espécies Reativas de Oxigênio/metabolismo
13.
Onco Targets Ther ; 12: 4585-4593, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31354292

RESUMO

PURPOSE: Pancreatic carcinoma is one of the most deadliest types of cancer, and relatively insensitive to the currently available chemotherapy. Thus, the discovery of novel therapeutic agents to prolong the survival times of patients with pancreatic cancer is urgently required. METHODS: Cell proliferation was assessed using the sulforhodamine B and cell clone formation assay, apoptosis was analyzed through Annexin V/PI staining, analysis of cell cycle distribution was determined by PI staining, and the expression of proteins was detected via Western blotting. RESULTS: Our data showed that harmine exerted an anti-proliferative effect and cell cycle arrest at G2/M in pancreatic cancer cells. Meanwhile, harmine plus gemcitabine showed strong synergy in inhibiting the proliferation of pancreatic cancer cells. Furthermore, harmine induced apoptosis and enhanced the gemcitabine-induced apoptosis in pancreatic cancer cells. The AKT/mTOR pathway is involved in mechanisms of gemcitabine resistance in pancreatic cancer cells, our data demonstrated that harmine plus gemcitabine significantly suppressed the AKT/mTOR signaling pathway. CONCLUSION: Harmine may be a potential candidate for the treatment of pancreatic cancer. Morever, the combination of harmine with gemcitabine appears to be an attractive option for the treatment of patients with pancreatic cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA