Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Macromol Rapid Commun ; 44(20): e2300350, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37535659

RESUMO

Nonfullerene acceptors with mediate bandgap play a crucial role in ternary devices as the third component, further boosting the performance of organic solar cells (OSCs). Herein, three F-series acceptors (F-H, F-Cl, and F-2Cl) with mediate bandgap are selected and introduced into the PM6:BDT-Br binary system as third component to find the detailed influence of end groups with chlorine (Cl) atom substitution on the performance of ternary organic solar cells. Due to the increased substitution of Cl atoms on the end groups, F-Cl and F-2Cl as guest acceptors reveal a superior ability to regulate the morphology of blend films, contributing to the ordered packing properties and high crystallinity. As a result, F-Cl and F-2Cl based ternary OSCs achieve significantly improved PCEs of 13.89% and 14.67%, respectively, compared with the binary devices (12.70%). On the contrary, F-H without Cl atom displays a poor compatibility with the host system, resulting in an inferior ternary device with a low PCE of 10.79%. This work indicates that F-series acceptors with mediate bandgap are a promising class of third component for high-performance ternary OSCs. And introducing more Cl atoms substitution on the end groups, especially F-2Cl, will own a broad applicability for other binary devices.


Assuntos
Bandagens , Halogênios , Eritromicina
2.
Am J Pathol ; 192(6): 970-983, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35358472

RESUMO

Lipid metabolic abnormalities in cancer cells are increasingly being studied. Several studies have reported that phosphatidylserine-specific phospholipase A1 (PLA1A) might be involved in the pathogenesis of cancers. Nevertheless, the function and mechanistic details of PLA1A in lung adenocarcinoma (LUAD) progression remain largely undefined. In the present study, low PLA1A expression was correlated with poor prognosis in patients with LUAD. Results from in vitro and in vivo animal studies showed that overexpressed PLA1A suppressed the proliferation of LUAD cells in vitro and tumor growth in vivo through regulation of cyclin abundance, thereby inducing S-phase arrest. Meanwhile, PLA1A overexpression attenuated migration and invasion of LUAD cells, including by inhibiting the epithelial-mesenchymal transition. Mechanistically, PLA1A overexpression inhibited aggressiveness of LUAD cells through elevated lysophosphatidylserine, which acts via G-protein-coupled receptor 174, further activating cAMP/protein kinase A pathway. Activating G-protein-coupled receptor 174/protein kinase A pathway may involve effects on cell cycle regulators and transcription factors-regulated epithelial-mesenchymal transition. The study uncovered the mechanism through which PLA1A regulates LUAD proliferation, invasion, and migration. These results demonstrate the potential use of PLA1A as a biomarker for diagnosing LUAD, which may therefore potentially serve as a therapeutic target for LUAD.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Adenocarcinoma de Pulmão/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Lisofosfolipídeos , Fosfatidilserinas , Fosfolipases A1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
3.
Clin Epigenetics ; 13(1): 220, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34906185

RESUMO

BACKGROUND: Early lung cancer detection remains a clinical challenge for standard diagnostic biopsies due to insufficient tumor morphological evidence. As epigenetic alterations precede morphological changes, expression alterations of certain imprinted genes could serve as actionable diagnostic biomarkers for malignant lung lesions. RESULTS: Using the previously established quantitative chromogenic imprinted gene in situ hybridization (QCIGISH) method, elevated aberrant allelic expression of imprinted genes GNAS, GRB10, SNRPN and HM13 was observed in lung cancers over benign lesions and normal controls, which were pathologically confirmed among histologically stained normal, paracancerous and malignant tissue sections. Based on the differential imprinting signatures, a diagnostic grading model was built on 246 formalin-fixed and paraffin-embedded (FFPE) surgically resected lung tissue specimens, tested against 30 lung cytology and small biopsy specimens, and blindly validated in an independent cohort of 155 patients. The QCIGISH diagnostic model demonstrated 99.1% sensitivity (95% CI 97.5-100.0%) and 92.1% specificity (95% CI 83.5-100.0%) in the blinded validation set. Of particular importance, QCIGISH achieved 97.1% sensitivity (95% CI 91.6-100.0%) for carcinoma in situ to stage IB cancers with 100% sensitivity and 91.7% specificity (95% CI 76.0-100.0%) noted for pulmonary nodules with diameters ≤ 2 cm. CONCLUSIONS: Our findings demonstrated the diagnostic value of epigenetic imprinting alterations as highly accurate translational biomarkers for a more definitive diagnosis of suspicious lung lesions.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Pulmonares/diagnóstico , Nódulos Pulmonares Múltiplos/genética , Idoso , Biomarcadores Tumorais/análise , Metilação de DNA/genética , Detecção Precoce de Câncer/métodos , Detecção Precoce de Câncer/normas , Detecção Precoce de Câncer/estatística & dados numéricos , Epigênese Genética/genética , Feminino , Impressão Genômica/genética , Impressão Genômica/fisiologia , Humanos , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , Nódulos Pulmonares Múltiplos/etiologia
4.
Int J Chron Obstruct Pulmon Dis ; 16: 2817-2832, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34675506

RESUMO

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a major health problem associated with high mortality worldwide. Cigarette smoke (CS) exposure is the main cause of COPD. Glioma pathogenesis-related protein 1 (GLIPR1) plays a key role in cell growth, proliferation, and invasion; however, the role of GLIPR1 in COPD remains unclear. METHODS: To clarify the involvement of GLIPR1 in COPD pathogenesis, Glipr1 knockout (Glipr1-/-) mice were generated. Wild-type (WT) and Glipr1-/- mice were challenged with CS for 3 months. To illustrate how GLIPR1 regulates CS-induced airway damage, knockdown experiments targeting GLIPR1 and PLAU, as well as overexpression experiments of PLAU, were performed with human bronchial epithelial cells. RESULTS: Compared with WT mice, Glipr1-/- mice showed exacerbated CS-induced airway damage including lung inflammation, airway wall thickening, and alveolar destruction. After CS exposure, total proteins, total white cells, neutrophils, lymphocytes, IL-6, and matrix metalloproteinase-9 increased significantly in lung of Glipr1-/- mice than those in lung of WT mice. Furthermore, in vivo and in vitro experiments demonstrated that silencing of GLIPR1 inactivated PLAU/EGFR signaling and promoted caspase-1-dependent pyroptosis (a mode of inflammatory cell death) induced by CS and CS extract exposure, respectively. In vitro experiments further revealed the interaction between GLIPR1 and PLAU, and silencing of PLAU blocked EGFR signaling and promoted pyroptosis, while overexpression of PLAU activated EGFR signaling and reversed pyroptosis. CONCLUSION: To conclude, GLIPR1 played a pivotal role in COPD pathogenesis and protected against CS-induced inflammatory response and airway damage, including cell pyroptosis, through the PLAU/EGFR signaling. Thus, GLIPR1 may play a potential role in COPD treatment.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana , Doença Pulmonar Obstrutiva Crônica , Animais , Receptores ErbB/genética , Inflamação/genética , Inflamação/prevenção & controle , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Camundongos , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/prevenção & controle , Fumaça/efeitos adversos , Fumar/efeitos adversos , Ativador de Plasminogênio Tipo Uroquinase
5.
Nanomaterials (Basel) ; 11(10)2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34685207

RESUMO

Lithium-ion capacitors (LICs) are attracting increasing attention because of their potential to bridge the electrochemical performance gap between batteries and supercapacitors. However, the commercial application of current LICs is still impeded by their inferior energy density, which is mainly due to the low capacity of the cathode. Therefore, tremendous efforts have been made in developing novel cathode materials with high capacity and excellent rate capability. Graphene-based nanomaterials have been recognized as one of the most promising cathodes for LICs due to their unique properties, and exciting progress has been achieved. Herein, in this review, the recent advances of graphene-based cathode materials for LICs are systematically summarized. Especially, the synthesis method, structure characterization and electrochemical performance of various graphene-based cathodes are comprehensively discussed and compared. Furthermore, their merits and limitations are also emphasized. Finally, a summary and outlook are presented to highlight some challenges of graphene-based cathode materials in the future applications of LICs.

6.
Metabolism ; 122: 154823, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34197875

RESUMO

Hepatic gluconeogenesis plays a crucial role in maintaining blood glucose homeostasis in mammals. Globe knockout of suppressor of cytokine signalling-2 (SOCS2), a feedback inhibitor of cytokine signalling, has been shown resistant to high-fat-diet (HFD)-induced hepatic steatosis with impaired glucose tolerance in mice. However, the underlying mechanism of SOCS2 regulates hepatic glucose homeostasis still undefined. In the present study, we demonstrated that the hepatic SOCS2 expression is markedly reduced in fasted C57BL/6 J mice or db/db mice. Moreover, hepatic SOCS2 expression levels are induced by metformin treatment. Ablation of SOCS2 attenuates suppressing effects of metformin on gluconeogenesis in hepatocytes. Gain- and loss-of-function studies indicated that SOCS2 regulates hepatic gluconeogenic genes expression and glucose output by mediating JAK2/STAT5 signalling pathway in db/db mice. Mechanistically, we observed that SOCS2 inactivates STAT5 by attenuating the interaction between JAK2 and STAT5, which in turn reduces hepatic gluconeogenesis. The present study reveals a critical role of SOCS2 in regulating hepatic gluconeogenesis. The inhibitory effect of metformin on gluconeogenesis is mediated, at least in part, by upregulating SOCS2 and therefore reducing hepatic gluconeogenic genes expression. SOCS2 may represent a new therapeutic target for the treatment of diabetes.


Assuntos
Gluconeogênese/fisiologia , Hiperglicemia/metabolismo , Janus Quinase 2/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/fisiologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Citocinas/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Jejum/fisiologia , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Gluconeogênese/efeitos dos fármacos , Células HEK293 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hiperglicemia/tratamento farmacológico , Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos
7.
Stem Cell Res Ther ; 12(1): 216, 2021 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-33781349

RESUMO

BACKGROUND: Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are syndromes of acute respiratory failure with extremely high mortality and few effective treatments. Mesenchymal stem cells (MSCs) may reportedly contribute to tissue repair in ALI and ARDS. However, applications of MSCs have been restricted due to safety considerations and limitations in terms of large-scale production and industrial delivery. Alternatively, the MSC secretome has been considered promising for use in therapeutic approaches and has been advanced in pre-clinical and clinical trials. Furthermore, the MSC secretome can be freeze-dried into a stable and ready-to-use supernatant lyophilized powder (SLP) form. Currently, there are no studies on the role of MSC SLP in ALI. METHODS: Intratracheal bleomycin was used to induce ALI in mice, and intratracheal MSC SLP was administered as a treatment. Histopathological assessment was performed by hematoxylin and eosin, immunohistochemistry, and immunofluorescence staining. Apoptosis, inflammatory infiltration, immunological cell counts, cytokine levels, and mRNA- and protein-expression levels of relevant targets were measured by performing terminal deoxynucleotidyl transferase dUTP nick-end labeling assays, determining total cell and protein levels in bronchoalveolar lavage fluids, flow cytometry, multiple cytokine-detection techniques, and reverse transcriptase-quantitative polymerase chain reaction and western blot analysis, respectively. RESULTS: We found that intratracheal MSC SLP considerably promoted cell survival, inhibited epithelial cell apoptosis, attenuated inflammatory cell recruitment, and reversed immunological imbalances induced by bleomycin. MSC SLP inhibited the interleukin 6-phosphorylated signal transducer and activator of transcription signaling pathway to activate tumor protein 63-jagged 2 signaling in basal cells, suppress T helper 17 cell differentiation, promote p63+ cell proliferation and lung damage repair, and attenuate inflammatory responses. CONCLUSIONS: MSC SLP ameliorated ALI by activating p63 and promoting p63+ cell proliferation and the repair of damaged epithelial cells. The findings of this study also shed insight into ALI pathogenesis and imply that MSC SLP shows considerable therapeutic promise for treating ALI and ARDS.


Assuntos
Lesão Pulmonar Aguda , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/terapia , Animais , Interleucina-6/genética , Lipopolissacarídeos , Pulmão , Camundongos , Pós
8.
Clin Exp Pharmacol Physiol ; 48(4): 498-507, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33462866

RESUMO

Particulate matter (PM2.5) and cigarette smoke exposure are leading factors contributing to various diseases, especially respiratory diseases. The purpose of this research was to study the effects of PM2.5 and cigarette smoke on glycerol kinase 5 (GK5) expression and the possible mechanisms by which GK5 participates in lipid droplet (LD) synthesis in alveolar epithelial A549 cells. Real-time polymerase chain reaction (RT-PCR) and western blotting have been used for the detection of messenger RNA (mRNA) and protein expression respectively. GK5 overexpressing cells were established by lentivirus transfection, whereby lentiviral vectors deliver the gene into chromosomes, allowing stable expression. Affymetrix microarray analysis, a widely used tool for measuring genome-wide gene expression, has been used to explore differential gene expression profiles. A549 cells stimulated with PM2.5 and cigarette smoke extract (CSE) showed elevated GK5 expression in a dose-dependent manner. Transmission electron microscopy and oil red O staining were used to observe LDs in cells. Further, GK5 overexpressing cells showed increased LDs and upregulation of genes and proteins related to lipogenesis and lipid transportation. Affymetrix microarray analysis revealed that GK5 overexpression resulted in the differential expression of more than 109 genes, which were mainly involved in the regulation of cell death, cell survival, cellular movement and migration, and those involved in cellular growth and proliferation pathways. Overall, this study demonstrates that GK5 is upregulated during PM2.5 and cigarette smoke exposure and induces LD synthesis.


Assuntos
Glicerol Quinase , Material Particulado , Células A549 , Apoptose , Humanos , Gotículas Lipídicas , Doença Pulmonar Obstrutiva Crônica , Fumaça
9.
Respir Physiol Neurobiol ; 281: 103497, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32683072

RESUMO

The aim of this study was to investigate the role and mechanism of perilipin 2 (PLIN2) in Pseudomonas aeruginosa pulmonary infection in vivo and in vitro. Twenty-eight-week-old male Balb/c mice were randomly divided into the control and PAO1 (P. aeruginosa standard strain) groups, which were administered phosphate-buffered saline (PBS) or PAO1 intratracheal instillation, respectively. RAW264.7 cells and BEAS-2B cells were stimulated with PBS or PAO1. The mRNA levels of PLIN2 and cytosolic phospholipase A2 (cPLA2) were detected by PCR. The protein expression of PLIN2 was detected by WB. BEAS-2B cells were transfected with shRNA against PLIN2, and cell proliferation was measured by CCK8. After 72 h, the expression of the PLIN2 gene in the PAO1 group was significantly higher than that in the control group. Compared with those in the control group, the protein levels of PLIN2, cyclooxygenase-2 (COX-2), and nuclear transcription factor-κB (NF-κB) in the PAO1 group were upregulated, while the expression of PLIN5 protein was downregulated. Furthermore, lung injury in the PAO1 group was more severe than that in the control group, and the lipid droplet (LD) level in lung tissue of PAO1 mice increased. After stimulation with P. aeruginosa, PLIN2 and cPLA2 genes in RAW264.7 and BEAS-2B cells were upregulated. CCK8 assay showed that proliferation decreased significantly in cells transfected with shRNA against PLIN2. In mice infected with P. aeruginosa, LDs accumulated in the lung tissue through an increase in PLIN2, which may result in increased COX-2-mediated anti-inflammatory cytokine release. This study provides a new understanding of the mechanism of lung infection and a new target for the treatment of clinical pulmonary injury.


Assuntos
Gotículas Lipídicas/metabolismo , Pneumopatias/metabolismo , Perilipina-2/metabolismo , Infecções por Pseudomonas/metabolismo , Células RAW 264.7/metabolismo , Mucosa Respiratória/metabolismo , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosfolipases A2 Citosólicas/metabolismo , Pseudomonas aeruginosa
10.
J Thorac Dis ; 12(3): 712-723, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32274137

RESUMO

BACKGROUND: Tyrosine kinase inhibitors (TKIs), such as gefitinib, are widely used as standard treatments for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations. However, the subsequent inevitable drug resistance has become a major challenge in clinical treatment. The aim of this study was to investigate the role of tissue-type plasminogen activator (PLAT) in gefitinib resistance in NSCLC. METHODS: The function of PLAT was determined using gefitinib-resistant cells and a nude mouse model. The gene knockdown was achieved by Lentivirus based RNA silence technique. Expression of relevant genes and proteins, cell viability, proliferation, apoptosis, cell cycle, reactive oxygen species levels, mitochondrial membrane potential and differential gene expression was detected by RT-qPCR, western blot, cell counting kit-8 assay, EdU incorporation, flow cytometry, JC-1 dye assay and complementary DNA arrays. The effects of PLAT knockdown on tumorigenesis was analyzed in vivo. RESULTS: Gefitinib-resistant cells expressed higher levels of PLAT and that knockdown of PLAT in resistant cells restored gefitinib sensitivity. Tumor proliferation was limited in vivo following PLAT knockdown. Moreover, PLAT knockdown affected mitochondrial function, caused caspase activation and cell cycle arrest, and activated TNF-α signaling, leading to apoptosis of gefitinib-resistant PC9 cells. CONCLUSIONS: Our results suggest that PLAT reduces apoptosis of NSCLC cells and knockdown of PLAT enhances anticancer effect of gefitinib by upregulating TNF-α signaling.

11.
Chem Asian J ; 14(15): 2588-2593, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31157511

RESUMO

A new role for the Nazarov reagent was revealed in the [4+2] benzannulation reaction, and delivered a series of polyfunctional benzenes in high yields under mild conditions. This metal-free process not only expanded the application of the Nazarov reagent in organic synthesis, but also provided a new, efficient way to construct polyfunctional benzenes.

12.
J Exp Clin Cancer Res ; 38(1): 96, 2019 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-30791926

RESUMO

BACKGROUND: Drug resistance is common in cancer chemotherapy. This study investigates the role of Glycerol kinase 5 (GK5) in mediating gefitinib resistance in NSCLC. METHODS: The exosomal mRNA of GK5 was detected using a tethered cationic lipoplex nanoparticle (TCLN) biochip. Real-time PCR and Western blot were used to examine the expression of GK5 mRNA and protein in gefitinib-sensitive and -resistant human lung adenocarcinoma cells. The cell counting kit-8, EdU assay, flow cytometry, and JC-1 dye were used to measure cell proliferation, cell cycle, and the mitochondrial membrane potential. RESULTS: We found that the exosomal mRNA of GK5 in the plasma of patients with gefitinib-resistant adenocarcinoma was significantly higher compared with that of gefitinib-sensitive patients. The mRNA and protein levels of GK5 were significantly upregulated in gefitinib-resistant human lung adenocarcinoma PC9R and H1975 cells compared with gefitinib-sensitive PC9 cells. Silencing GK5 in PC9R cells induced mitochondrial damage, caspase activation, cell cycle arrest, and apoptosis via SREBP1/SCD1 signaling pathway. CONCLUSIONS: We demonstrated that GK5 confers gefitinib resistance in lung cancer by inhibiting apoptosis and cell cycle arrest. GK5 could be a novel therapeutic target for treatment of NSCLC with resistance to EGFR tyrosine kinase inhibitors.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Gefitinibe/farmacologia , Glicerol Quinase/genética , Transdução de Sinais/genética , Estearoil-CoA Dessaturase/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma de Pulmão/dietoterapia , Adenocarcinoma de Pulmão/genética , Animais , Antineoplásicos/farmacologia , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Masculino , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Endogâmicos BALB C , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , RNA Mensageiro/genética , Regulação para Cima/genética
13.
J Cancer Res Clin Oncol ; 145(4): 861-872, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30661098

RESUMO

PURPOSE: Non-small-cell lung cancer (NSCLC) is the most common form of lung cancer. Gefitinib is one of the most accepted therapies against NSCLC in those carrying EGFR mutations, but it is only effective in approximately 20% of patients with NSCLC. Thus, alternative therapeutic interventions are urgently needed to overcome gefitinib resistance. Glutaredoxin (GLRX) plays a key role in oxidative stress. However, whether GLRX inhibition could enhance gefitinib efficacy in the gefitinib-resistant NSCLC cells is unknown. In this study, we aimed to determine whether combined inhibition of GLRX could enhance growth-inhibitory effects of gefitinib in gefitinib-resistant NSCLC cells. METHODS: Real-time PCR and western blotting were used to examine the mRNA and protein levels of GLRX in gefitinib-sensitive PC9 and HCC827 and -resistant human lung adenocarcinoma PC9R, HCC827R, and H1975 cells. Cell Counting Kit-8, flow cytometry, JC-1 staining, and reactive oxygen species (ROS) assays were used to evaluate cell proliferation, cell cycle progression, mitochondrial membrane potential, and ROS generation, respectively. Mouse tumor xenografts were used to assess the effect of GLRX in vivo. RESULTS: We found that GLRX was upregulated in gefitinib-resistant PC9R, HCC827R, and H1975 cells. GLRX inhibition enhanced the effects of geftinib in gefitinib-resistant cell proliferation in vitro and in vivo and promoted apoptosis and cell cycle arrest via the EGFR/Forkhead Box M1 (FoxM1) signaling pathway, indicating that combined inhibition of GLRX could enhance growth-inhibitory effects of gefitinib in gefitinib-resistant NSCLC cells. CONCLUSIONS: Our results suggest that GLRX inhibition enhances the effects of geftinib in EGFR-TKI-resistant NSCLC cells. Thus, GLRX may represent a therapeutic target for increasing the efficiency of gefitinib treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proteína Forkhead Box M1/metabolismo , Gefitinibe/farmacologia , Glutarredoxinas/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glutarredoxinas/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Regulação para Cima
14.
Oncol Rep ; 40(4): 1937-1946, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30106446

RESUMO

Gefitinib, an epidermal growth factor receptor (EGFR)­specific drug, is effective for ~1 year, after which resistance is inevitable. Calpain 2 (CAPN2) is known to serve a role in the drug response and resistance in certain cancer therapies. However, the full function of CAPN2, particularly in non­small cell lung cancer, has not yet been elucidated. In the present study, CAPN2 expression in gefitinib­resistant lung adenocarcinoma cells was investigated. CAPN2 function in these cells was further evaluated using gene knockdown both in vitro and in vivo. The results demonstrated that CAPN2 was strongly associated with gefitinib­resistance, and CAPN2 mRNA and protein expression levels were significantly increased in gefitinib­resistant cell lines. Furthermore, CAPN2 knockdown inhibited gefitinib­resistant cell proliferation in vitro and in vivo. CAPN2 conferred gefitinib­resistance by inhibiting cell apoptosis and arresting the cell cycle. CAPN2 knockdown also induced caspase activation and mitochondrial dysfunction, and its function in gefitinib resistance appeared to be largely mediated by EGFR/protein kinase B/survivin signaling pathway activation. These results suggest that CAPN2 is responsible for EGFR­tyrosine kinase inhibitor resistance, and CAPN2 inhibition may be used to provide therapeutic benefits in the treatment of gefitinib resistance.


Assuntos
Apoptose/efeitos dos fármacos , Calpaína/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Calpaína/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/genética , Gefitinibe , Humanos , Proteínas Inibidoras de Apoptose/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais , Survivina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cell Physiol Biochem ; 47(5): 1909-1924, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29961070

RESUMO

BACKGROUND/AIMS: Tyrosine kinase inhibitor gefitinib significantly improves the survival of patients with non-small-cell lung cancer (NSCLC) by inhibiting epidermal growth factor receptor (EGFR) tyrosine kinase. However, patients eventually develop resistance to gefitinib through uncharacterized mechanisms. It is known that plasminogen activator urokinase receptor (PLAUR) plays an important role in cell proliferation, migration and apoptosis. However, the role of PLAUR, particularly exosomal PLAUR in gefitinib resistance in NSCLC has not been reported. The aim of this study is to determine the relationship between PLAUR and gefitinib resistance. METHODS: In this study, a tethered cationic lipoplex nanoparticle (TCLN) biochip containing molecular beacons was used as probes to detect PLAUR mRNA in plasma exosomes from patients with gefitinib-sensitive and -resistant NSCLC. In vitro, Real-time PCR was used to examine the expression of PLAUR mRNA and Western blot was applied to examine the expression of related proteins. The gene knockdown was achieved by Lentivirus based RNA silence technique. The cell counting kit-8 assay and EdU incorporation were used to examine cell proliferation. The flow cytometry was applied to determine cell apoptosis and cell cycle, while the mitochondrial membrane potential was measured by JC-1 dye assay. Signaling pathway affected by PLAUR knockdown was identified by cDNA Microarray. The effect of PLAUR knockdown on tumorigenesis was analyzed in vivo. RESULTS: We found that the exosomal PLAUR mRNA in the plasma of gefitinib-resistant NSCLC patients was significantly increased compared to that of gefitinib-sensitive NSCLC patients. The PLAUR mRNA and soluble PLAUR protein were also significantly increased in gefitinib-resistant human lung adenocarcinoma PC9R cells compared to gefitinib-sensitive PC9 cells. Silencing PLAUR in PC9R cells impaired mitochondrial membrane potential and increased cell apoptosis via EGFR/p-AKT/survivin signaling pathway. Furthermore, EGFR was upregulated in the geftinib-resistant PC9R cells, and knockdown of EGFR significantly increased cell apoptosis. CONCLUSIONS: Taken together, our results demonstrated that PLAUR induces geftinib-resistance through EGFR/p-AKT/survivin signaling pathway in gefitinib-resistant human lung adenocarcinoma cells. PLAUR could be a novel therapeutic target for gefitinib-resistant NSCLC patients.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Receptores ErbB/genética , Feminino , Gefitinibe , Humanos , Proteínas Inibidoras de Apoptose/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transdução de Sinais/genética , Survivina
16.
Respir Res ; 19(1): 96, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29784046

RESUMO

BACKGROUND: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, including gefitinib, are first-line drugs against advanced non-small cell lung cancer with activating EGFR mutations. However, the development of resistance to such drugs is a major clinical challenge. METHODS: The role of annexin A5 in resistance to EGFR tyrosine kinase inhibitors was investigated by qPCR and western blot of relevant molecules, by CCK8 and EdU assay of cell proliferation and viability, by annexin V/propidium iodide assay of apoptosis and cell cycle distribution, by JC-1 assay of mitochondrial integrity, and by xenograft assay of tumorigenicity. RESULTS: We found that annexin A5 is upregulated in gefitinib-resistant cell lines, as well as in clinical specimens resistant to EGFR tyrosine kinase inhibitors. Accordingly, knockdown of the gene from gefitinib-resistant cells restores gefitinib sensitivity in vitro and in vivo by downregulating polo-like kinase 1 signal pathway, thereby inducing mitochondrial damage, caspase activation, cell cycle arrest at G2/M, and, finally, apoptosis. CONCLUSIONS: The data indicate that annexin A5 confers gefitinib resistance in lung cancer by inhibiting apoptosis and G2/M cell cycle arrest, and is thus a potential therapeutic target in non-small cell lung cancers resistant to EGFR tyrosine kinase inhibitors.


Assuntos
Anexina A5/deficiência , Antineoplásicos/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/fisiologia , Técnicas de Silenciamento de Genes , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Quinazolinas/farmacologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma de Pulmão , Animais , Anexina A5/biossíntese , Anexina A5/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Gefitinibe , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus
17.
J Huazhong Univ Sci Technolog Med Sci ; 36(3): 463-468, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27376821

RESUMO

The human pregnane X receptor (hPXR) plays a critical role in the metabolism, transport and clearance of xenobiotics in the liver and intestine. The hPXR can be activated by a structurally diverse of drugs to initiate clinically relevant drug-drug interactions. In this article, in silico investigation was performed on a structurally diverse set of drugs to identify critical structural features greatly related to their agonist activity towards hPXR. Heuristic method (HM)-Best Subset Modeling (BSM) and HM-Polynomial Neural Networks (PNN) were utilized to develop the linear and non-linear quantitative structure-activity relationship models. The applicability domain (AD) of the models was assessed by Williams plot. Statistically reliable models with good predictive power and explain were achieved (for HM-BSM, r (2)=0.881, q LOO (2) =0.797, q EXT (2) =0.674; for HM-PNN, r (2)=0.882, q LOO (2) =0.856, q EXT (2) =0.655). The developed models indicated that molecular aromatic and electric property, molecular weight and complexity may govern agonist activity of a structurally diverse set of drugs to hPXR.


Assuntos
Modelos Estatísticos , Relação Quantitativa Estrutura-Atividade , Receptores de Esteroides/agonistas , Bibliotecas de Moléculas Pequenas/química , Simulação por Computador , Humanos , Peso Molecular , Redes Neurais de Computação , Receptor de Pregnano X , Receptores de Esteroides/química , Eletricidade Estática
18.
Chem Asian J ; 11(10): 1512-7, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-26990982

RESUMO

A new strategy for the one-pot synthesis of polysubstituted benzenes through a N,N-dimethyl-4-aminopyridine (DMAP)-catalyzed [4+2] benzannulation from readily prepared 1,3-bis(sulfonyl)butadienes and γ-substituted allenoates is described. This method provides a facile, metal-free and general route to highly substituted benzenes under mild conditions in moderate-to-good yields with complete regioselectivity.


Assuntos
Aminopiridinas/química , Benzoatos/síntese química , Butadienos/química , Catálise , Ciclização , Modelos Químicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...