Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Invest Dermatol ; 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38580105

RESUMO

IL-6 signaling plays a crucial role in the survival and metastasis of skin cancer. NEDD4L acts as a suppressor of IL-6 signaling by targeting GP130 degradation. However, the effects of the NEDD4L-regulated IL-6/GP130 signaling pathway on skin cancer remain unclear. In this study, protein expression levels of NEDD4L and GP130 were measured in tumor tissues from patients with cutaneous squamous cell carcinoma. Skin tumors were induced in wild-type and Nedd4l-knockout mice, and activation of the IL-6/GP130/signal transducer and activator of transcription 3 signaling pathway was detected. The results indicated a negative correlation between the protein expression levels of NEDD4L and GP130 in cutaneous squamous cell carcinoma tissues from patients. Nedd4l deficiency significantly promoted 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol-13-acetate-induced skin tumorigenesis and benign-to-malignant conversion by activating the IL-6/GP130/signal transducer and activator of transcription 3 signaling pathway, which was abrogated by supplementation with the GP130 inhibitor SC144. Furthermore, our findings suggested that NEDD4L can interact with GP130 and promote its ubiquitination in skin tumors. In conclusion, our results indicate that NEDD4L could act as a tumor suppressor in skin cancer, and inhibition of GP130 could be a potential therapeutic method for treating this disease.

2.
Lung ; 200(5): 619-631, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36107242

RESUMO

PURPOSE: It has been shown that activation of autophagy promotes the development of pulmonary arterial hypertension (PAH). Meanwhile, forkhead box M1 (FOXM1) has been found to induce autophagy in several types of cancer. However, it is still unclear whether FOXM1 mediates autophagy activation in PAH, and detailed mechanisms responsible for these processes are indefinite. METHOD: PAH was induced by a single intraperitoneal injection of monocrotaline (MCT) to rats. The right ventricle systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), percentage of medial wall thickness (%MT), α-smooth muscle actin (α-SMA) staining, and Ki67 staining were performed to evaluate the development of PAH. The protein levels of FOXM1, phospho-focal adhesion kinase (p-FAK), FAK, and LC3B were determined by immunoblotting or immunohistochemistry. RESULTS: FOXM1 protein level and FAK activity were significantly increased in MCT-induced PAH rats, this was accompanied with the activation of autophagy. Pharmacological inhibition of FOXM1 or FAK suppressed MCT-induced autophagy activation, decreased RVSP, RVHI and %MT in MCT-induced PAH rats, and inhibited the proliferation of pulmonary arterial smooth muscle cells and pulmonary vessel muscularization in MCT-induced PAH rats. CONCLUSION: FOXM1 promotes the development of PAH by inducing FAK phosphorylation and subsequent activation of autophagy in MCT-treated rats.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Actinas/metabolismo , Animais , Autofagia , Modelos Animais de Doenças , Hipertensão Pulmonar Primária Familiar , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/metabolismo , Antígeno Ki-67/metabolismo , Monocrotalina/metabolismo , Monocrotalina/toxicidade , Fosforilação , Hipertensão Arterial Pulmonar/induzido quimicamente , Artéria Pulmonar , Ratos , Ratos Sprague-Dawley
3.
Pulm Circ ; 11(4): 20458940211046131, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34552711

RESUMO

Pulmonary arterial hypertension is a devastating pulmonary vascular disease, in which the pathogenesis is complicated and unclear. Pulmonary arterial smooth muscle cells (PASMCs) proliferation is a key pathological feature of pulmonary arterial hypertension. It has been shown that ubiquitin-specific protease 7 (USP7) is involved in cancer cell proliferation via deubiquitinating and stabilizing E3 ubiquitin ligase mouse double minute 2 (MDM2). However, the effect of USP7 and MDM2 on platelet-derived growth factor (PDGF)-induced PASMCs proliferation is uncertain. This study aims to explore this issue. Our results indicated that PDGF up-regulated USP7 protein expression and stimulated PASMCs proliferation; this was accompanied with the increase of MDM2, forkhead box O4 (FoxO4) reduction and elevation of CyclinD1. While prior transfection of USP7 siRNA blocked PDGF-induced MDM2 up-regulation, FoxO4 down-regulation, increase of CyclinD1 and cell proliferation. Pre-depletion of MDM2 by siRNA transfection reversed PDGF-induced reduction of FoxO4, up-regulation of CyclinD1 and PASMCs proliferation. Furthermore, pre-treatment of cells with proteasome inhibitor MG-132 also abolished PDGF-induced FoxO4 reduction, CyclinD1 elevation and cell proliferation. Our study suggests that USP7 up-regulates MDM2, which facilitates FoxO4 ubiquitinated degradation, and subsequently increases the expression of CyclinD1 to mediate PDGF-induced PASMCs proliferation.

4.
Mol Cell Biochem ; 476(8): 3037-3049, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33797701

RESUMO

Galectin-3(Gal-3) is an effective regulator in the pathological process of pulmonary arterial hypertension (PAH). However, the detailed mechanisms underlying Gal-3 contribution to PAH are not yet entirely clear. The aim of the present study was to explore these issues. Proliferation of rat pulmonary arterial smooth muscle cells (PASMCs) was determined using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. Small interfering RNA (siRNA) was applied to silence the expression of yes-associated protein (YAP) and Forkhead box M1 (FOXM1). The protein expression and phosphorylation were measured by immunoblotting. The subcellular location of YAP was determined using immunoblotting and immunofluorescence. Gal-3-stimulated PASMCs proliferation in a time- and dose-dependent manner, this was accompanied with, YAP upregulation, dephosphorylation, and nucleus translocation. Gal-3 further increased FOXM1 and cyclinD1 expression via YAP activation. Interfering YAP/FOXM1 axis suppressed Gal-3-induced PASMCs proliferation. Activation of AMPK also inhibited Gal-3-triggered cells proliferation by targeting YAP/FOXM1/cyclinD1 pathway. Gal-3 induced PASMCs proliferation by regulating YAP/FOXM1/cyclinD1 signaling cascade, and activation of AMPK targeted on this axis and suppressed Gal-3-stimulated PASMCs proliferation. Our study provides novel therapeutic targets for prevention and treatment of PAH.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proliferação de Células , Galectina 3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miócitos de Músculo Liso/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Artéria Pulmonar/citologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Apoptose , Movimento Celular , Galectina 3/genética , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Miócitos de Músculo Liso/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas de Sinalização YAP
5.
Front Cell Infect Microbiol ; 11: 541092, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777827

RESUMO

Background: Metagenomic next-generation sequencing (mNGS) is a powerful method for pathogen detection. In this study, we assessed the value of mNGS for bronchoalveolar lavage (BAL) samples in the diagnosis of pulmonary infections. Methods: From February 2018 to April 2019, BAL samples were collected from 235 patients with suspected pulmonary infections. mNGS and microbial culture were performed to evaluate the effectiveness of mNGS in pulmonary infection diagnosis. Results: We employed mNGS to evaluate the alpha diversity, results suggesting that patients with confirmed pathogens had a lower microbial diversity index compared to that of patients with uncertain pathogens. For the patients admitted to the respiratory intensive care unit (RICU) or on a ventilator, they experienced a lower diversity index than that of the patients in the general ward or not on a ventilator. In addition, mNGS of BAL had a diagnostic sensitivity of 88.89% and a specificity of 14.86% in pulmonary infection, with 21.16% positive predictive value (PPV) and 83.87% negative predictive value (NPV). When rare pathogens were excluded, the sensitivity of mNGS decreased to 73.33%, and the specificity increased to 41.71%. For patients in the simple pulmonary infection group and the immunocompromised group, the main infection types were bacterial infection (58.33%) and mixed-infection (43.18%). Furthermore, mNGS had an advantage over culture in describing polymicrobial ecosystem, demonstrating the microbial distribution and the dominant strains of the respiratory tract in patients with different underlying diseases. Conclusions: The study indicated that mNGS of BAL samples could provide more accurate diagnostic information in pulmonary infections and demonstrate the changes of respiratory microbiome in different underlying diseases. This method might play an important role in the clinical use of antimicrobial agents in the future.


Assuntos
Ecossistema , Metagenômica , Lavagem Broncoalveolar , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Sensibilidade e Especificidade
6.
Am J Manag Care ; 27(2): e36-e41, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33577159

RESUMO

OBJECTIVES: This study aimed to evaluate factors affecting adherence to inhaled therapy in patients with asthma to further identify the determinants most closely associated with adherence to inhaled therapy for asthma, especially inhaled glucocorticosteroids (ICS). STUDY DESIGN: A 2-stage study was conducted. In stage 1, we performed nonassumptive deep-dive qualitative scoping to investigate the determinants of poor adherence in patients with asthma, and in stage 2 we developed a new questionnaire for cross-sectional surveys to obtain more accurate information about critical issues related to asthma management. METHODS: Patients with asthma who were 18 years and older in the outpatient clinic of The First Affiliated Hospital of Xi'an Jiaotong University from November 2016 to January 2018 were investigated. RESULTS: In the 350 patients with asthma recruited, 32% of patients showed good adherence, whereas 68% of patients displayed poor adherence to inhaled therapy due to various reasons. Further analysis indicated that inadequate understanding of asthma treatment and control, poor self-management, financial burden, adverse reactions, and the fear of potential adverse reactions were significant independent risk factors for poor ICS inhalation adherence in patients with asthma. CONCLUSIONS: Our research shows that many patients with asthma in western China have poor disease control and poor inhalation therapy adherence. We hope this research can alert clinicians and help them identify patients who may be experiencing uncontrolled asthma due to poor adherence to inhaled therapy, and we suggest that clinicians help those patients obtain appropriate information about asthma control and self-management.


Assuntos
Corticosteroides , Asma , Administração por Inalação , Corticosteroides/uso terapêutico , Asma/tratamento farmacológico , Estudos Transversais , Humanos , Adesão à Medicação
7.
J Cell Physiol ; 236(6): 4694-4708, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33283886

RESUMO

The aims of the present study were to examine the molecular mechanisms underlying sphingosine-1-phosphate (S1P)-induced rat pulmonary artery smooth muscle cells (PASMCs) proliferation/migration and to determine the effect of yes-associated protein (YAP) activation on S1P-induced PASMCs proliferation/migration and its potential mechanisms. S1P induced YAP dephosphorylation and nuclear translocation, upregulated microRNA-130a/b (miR-130a/b) expression, reduced bone morphogenetic protein receptor 2 (BMPR2), and inhibitor of DNA binding 1(Id1) expression, and promoted PASMCs proliferation and migration. Pretreatment of cells with Rho-associated protein kinase (ROCK) inhibitor Y27632 suppressed S1P-induced YAP activation, miR-130a/b upregulation, BMPR2/Id1 downregulation, and PASMCs proliferation/migration. Knockdown of YAP using small interfering RNA also suppressed S1P-induced alterations of miR-130a/b, BMPR2, Id1, and PASMCs behavior. In addition, luciferase reporter assay indicated that miR-130a/b directly regulated BMPR2 expression in PASMCs. Inhibition of miR-130a/b functions by anti-miRNA oligonucleotides attenuated S1P-induced BMPR2/Id1 downregulation and the proliferation and migration of PASMCs. Taken together, our study indicates that S1P induces activation of YAP through ROCK signaling and subsequently increases miR-130a/b expression, which, in turn, downregulates BMPR2 and Id1 leading to PASMCs proliferation and migration.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisofosfolipídeos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Esfingosina/análogos & derivados , Transporte Ativo do Núcleo Celular , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Cultivadas , Proteína 1 Inibidora de Diferenciação/metabolismo , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosforilação , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Esfingosina/farmacologia , Proteínas de Sinalização YAP , Quinases Associadas a rho/metabolismo
8.
Eur J Pharmacol ; 884: 173302, 2020 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-32659302

RESUMO

It has been shown that sphingosine-1-phosphate (S1P) is elevated in patients with pulmonary arterial hypertension (PAH) and promotes the proliferation of pulmonary artery smooth muscle cells (PASMCs). Meanwhile, S1P has been found to induce the activation of autophagy in several types of human diseases including cancers. However, it is still unclear whether activation of autophagy mediates S1P-induced PASMCs proliferation, and detailed mechanisms responsible for these processes are indefinite. The aims of this study are to address these issues. S1P dose- and time-dependently reduced the expression of E-cadherin/CDH1 and stimulated PASMCs proliferation; this was accompanied with the elevation of TNF receptor-associated factor 2 (TRAF2), up-regulation and ubiquitination of BECN1 and the activation of autophagy. Prior silencing TRAF2 or BECN1 using siRNA or pre-incubation of cells with autophagy inhibitor chloroquine phosphate (CQ) suppressed S1P-induced autophagy activation and subsequent CDH1 degradation and further PASMCs proliferation. Taken together, our study indicates that S1P promotes the activation of autophagy by accelerating TRAF2-mediated BECN1 up-regulation and ubiquitination, which in turn results in CDH1 reduction and contributes to PASMCs proliferation.


Assuntos
Autofagia/efeitos dos fármacos , Caderinas/metabolismo , Proliferação de Células/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Caderinas/genética , Células Cultivadas , Regulação para Baixo , Masculino , Músculo Liso Vascular/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Esfingosina/farmacologia , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Ubiquitinação
9.
BMC Pulm Med ; 20(1): 182, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32586317

RESUMO

BACKGROUND: In recent years, many studies have discovered that cystatin C (Cys C) may play an important role in respiratory diseases, especially in chronic obstructive pulmonary disease (COPD). However, the findings of these studies were inconsistent. This systematic review and meta-analysis aimed to assess the relationship between serum Cys C and COPD. METHODS: We conducted a systematic literature search in PubMed, Embase, Web of Science, Wanfang databases, and the China National Knowledge Infrastructure. The standardized mean difference (SMD), Fisher's Z-value and 95% confidence interval (CI) were calculated to investigate the effect sizes. Subgroup analyses were performed on disease status, ethnicity, assay method, and study design. Sensitivity was performed, and publication bias was assessed. RESULTS: A total of 15 studies, including 4079 COPD patients and 5949 controls, were included in this meta-analysis. The results showed that serum Cys C levels in patients with COPD were significantly higher than those in controls (SMD = 0.99, 95% CI =0.62-1.37, P < 0.001), especially in AECOPD (SMD = 1.59, 95% CI =1.05-2.13, P < 0.001), and there were statistically different among AECOPD and SCOPD (SMD = 0.35, 95% CI =0.10-0.59, P = 0.005). The serum Cys C levels were negatively correlated with FEV1%pre (Z = - 0.45, 95%CI = -0.58--0.32, P = 0.011) and FEV1/FVC (Z = - 0.32, 95%CI = -0.50--0.14, P = 0.006). The serum Cys C levels were independent of ethnicity, assay method, and study design. CONCLUSION: Serum Cys C levels were associated with COPD and COPD exacerbation, and they were inversely correlated with FEV1%pre and FEV1/FVC.


Assuntos
Cistatina C/sangue , Doença Pulmonar Obstrutiva Crônica/sangue , Doença Pulmonar Obstrutiva Crônica/patologia , Biomarcadores/sangue , Progressão da Doença , Humanos
10.
Life Sci ; 242: 117159, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-31837334

RESUMO

AIMS: It has been shown that up-regulation of E3 ubiquitin ligase seven-in-absentia-homolog 2 (Siah2) and activation of Hippo signaling pathway effector yes-associated protein (YAP) are involved in the development of pulmonary arterial hypertension (PAH). However, it is still unclear whether Siah2 activates YAP in monocrotaline (MCT)-induced PAH rat models. MAIN METHODS: Intraperitoneal injection of MCT was used to induce PAH rat models. The right ventricular systolic pressure (RVSP), right ventricle hypertrophy index (RVHI), percentage of medial wall thickness (%MT), α-SMA, Ki-67 and TUNEL staining were performed to evaluate the development of PAH. Protein levels of Siah2, Lats1/2, YAP phosphorylation and total YAP, and the subcellular localization of YAP were examined using immunoblotting. Proteasome activity was measured by an assay kit. KEY FINDINGS: The protein level of Siah2 was significantly increased in MCT-induced PAH rats, this was accompanied with the proteasome-dependent degradation of Lats1/2 and subsequent up-regulation and dephosphorylation of YAP and its nuclear localization. Administration of PAH rats with Siah2 inhibitor Vitamin K3 or proteasome inhibitor MG-132 dramatically suppressed MCT-induced down-regulation of Lats1/2 and activation of YAP, finally reduced RVSP, RVHI, %MT, pulmonary arterial muscularization, pulmonary arterial smooth muscle cells (PASMCs) proliferation and enhanced PASMCs apoptosis in PAH rats. SIGNIFICANCE: Siah2 contributes to the development of MCT-induced PAH by destabilizing Lats1/2 and subsequently stimulating YAP activation. Inhibition of Siah2 or proteasome alleviates pulmonary arterial remodeling through inactivation of YAP, indicating Siah2 ubiquitin ligase as a novel target might have potential value in the management of PAH.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Monocrotalina/farmacologia , Proteínas Nucleares/fisiologia , Hipertensão Arterial Pulmonar/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Remodelação Vascular/efeitos dos fármacos , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Immunoblotting , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Hipertensão Arterial Pulmonar/induzido quimicamente , Hipertensão Arterial Pulmonar/fisiopatologia , Ratos , Ratos Sprague-Dawley , Remodelação Vascular/fisiologia , Proteínas de Sinalização YAP
11.
Eur J Pharmacol ; 867: 172823, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31770525

RESUMO

Leukotriene B4 (LTB4) has been found to contribute to pulmonary arterial smooth muscle cells (PASMCs) proliferation and pulmonary arterial remodeling therefore the development of pulmonary arterial hypertension (PAH). Yet, the underlying molecular mechanisms remain poorly understood. The present study aims to address this issue. Our results demonstrate that LTB4 dose- and time-dependently induced proliferation of primary cultured rat PASMCs, this was accompanied with the activation of phosphatidylinositol-3-kinase/Akt (PI3K/Akt) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways, and consequent inactivation of glycogen synthase kinase-3ß (GSK-3ß), up-regulation of ß-catenin and induction of cyclin D1 expression. The presence of PI3K inhibitor (LY294002) or MEK inhibitor (U0126) or prior silencing of ß-catenin with siRNA suppressed LTB4-induced cyclin D1 up-regulation and PASMCs proliferation. In addition, inactivation or lack of GSK-3ß up-regulated ß-catenin and cyclin D1 in PASMCs. Taken together, our study indicates that activation of PI3K/Akt and ERK1/2 pathways mediates LTB4-induced PASMCs proliferation by modulating GSK-3ß/ß-catenin/cyclin D1 axis and suggests that targeting this pathway might have potential value in alleviating vascular remodeling and benefit PAH.


Assuntos
Hipertensão Pulmonar/imunologia , Leucotrieno B4/imunologia , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/patologia , Remodelação Vascular/imunologia , Animais , Butadienos/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Ciclina D1/imunologia , Ciclina D1/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Hipertensão Pulmonar/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Morfolinas/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/imunologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Cultura Primária de Células , Artéria Pulmonar/imunologia , Artéria Pulmonar/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Remodelação Vascular/efeitos dos fármacos , beta Catenina/genética , beta Catenina/metabolismo
12.
J Cancer ; 10(23): 5754-5763, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31737112

RESUMO

Background: A number of studies have investigated the role of Golgi phosphoprotein-3 (GOLPH3) in the pathogenesis and progression of non-small cell lung cancer (NSCLC). However, the results of previous studies are heterogeneous and controversial. The aim of this meta-analysis was to clarify its association with the clinicopathological characteristics of patients and evaluate the prognostic significance of GOLPH3 in NSCLC. Methods: A systematic search was conducted through PMC, PubMed, Medline, Web of Science, Chinese National Knowledge Infrastructure and Wanfang database. The odds ratio (OR) and hazard ratio (HR) with 95 % CI were calculated by STATA 12.0. Results: 8 qualified studies with a total of 1001 patients with NSCLC were included. Pooled results showed that GOLPH3 was highly expressed in tumor tissues compared with adjacent lung tissues (OR, 7.55), and overexpression of GOLPH3 was significantly correlated with advanced clinical stage (OR, 3.42), poor differentiation of tumor (OR, 1.97) and positive lymph node metastasis (OR, 2.58), but no association with histological type, gender, age or tumor size was found in NSCLC patients. In addition, the pooled HR for overall survival was 1.79 by univariate analysis and 1.91 by multivariate analysis. The pooled HR for progression-free survival was 2.50. Conclusions: GOLPH3 could be a risk factor for progression of NSCLC and might act as a potential prognostic biomarker for NSCLC patients.

13.
Artif Cells Nanomed Biotechnol ; 47(1): 3315-3321, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31385542

RESUMO

Objective: The long intergenic non-coding RNA 01296 (LINC01296) has been reported to be overexpressed in multiple tumours. However, the role of LINC01296 in clinicopathologic and prognostic value in cancers remains completely unknown. The aim of the present meta-analysis was to comprehensively elucidate the correlation between LINC01296 with clinicopathological features and survival outcomes in tumours. Methods: Electronic databases of PubMed, Web of Science, Chinese National Knowledge Infrastructure (CNKI), and Wanfang Database were used to search relevant studies. The role of LINC01296 in cancers was evaluated by pooled hazard ratios (HRs), odds ratios (ORs) and 95% confidence intervals (CIs). Results: In total, nine studies compromising 720 participants were enrolled in this analysis. The pooled results showed increased LINC01296 expression could predict unfavourable overall survival (OS) (HR = 1.89, 95%CI = 1.47-2.43, p < .001). Additionally, elevated LINC01296 expression was correlated with clinical stage (OR = 2.95, 95%CI = 2.13-4.08, p < .001), lymph node metastasis (OR = 2.76, 95%CI = 2.00-3.81, p < .001), tumour size (OR = 2.80, 95%CI = 1.77-4.41, p < .001), and tumour differentiation (OR = 2.11, 95%CI = 1.36-3.27, p < .001) in patients with cancers. Conclusion: The results of this meta-analysis indicated LINC01296 was a novel biomarker for prognosis and clinicopathological parameters in cancers.


Assuntos
Neoplasias/diagnóstico , Neoplasias/genética , RNA Longo não Codificante/genética , Humanos , Neoplasias/patologia , Prognóstico , Análise de Sobrevida
14.
Biochem Biophys Res Commun ; 516(3): 921-927, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31277946

RESUMO

The upregulation of osteopontin(OPN) has been found to contribute to the proliferation of pulmonary artery smooth muscle cells(PASMCs), and activation of PPARγ has been shown to suppress OPN expression in THP-1 cells. However, the molecular mechanisms underlying the upregulation of OPN expression and PPARγ agonist modulation of OPN expression in PASMCs remain largely unclear. Here we found that S1P stimulated PASMCs proliferation and up-regulated OPN expression in rat PASMCs, which was accompanied with the activation of phospholipase C(PLC), calcineurin and translocation of NFATc3 to nucleus. Further study showed that inhibition of PLC by U73122, suppression of calcineurin activity by cyclosporine A(CsA) or knockdown of NFATc3 using small interfering RNA suppressed S1P-induced OPN up-regulation. Activation of PPARγ by pioglitazone suppressed S1P-induced activation of calcineurin/NFATc3 signaling pathway and followed OPN up-regulation. Taken together, our study indicates that S1P stimulates OPN expression by activation of PLC/calcineurin/NFATc3 signaling pathway, and activation of PPARγ suppresses calcineurin/NFATc3-mediated OPN expression in PASMCs.


Assuntos
Calcineurina/genética , Lisofosfolipídeos/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fatores de Transcrição NFATC/genética , Osteopontina/genética , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Cátions Bivalentes , Proliferação de Células/efeitos dos fármacos , Ciclosporina/farmacologia , Estrenos/farmacologia , Feminino , Regulação da Expressão Gênica , Transporte de Íons , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Osteopontina/metabolismo , PPAR gama/antagonistas & inibidores , PPAR gama/genética , PPAR gama/metabolismo , Pioglitazona/farmacologia , Cultura Primária de Células , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Pirrolidinonas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Esfingosina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo
15.
Naunyn Schmiedebergs Arch Pharmacol ; 392(5): 605-613, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30683943

RESUMO

It has been demonstrated that activation of autophagy is involved in the development of pulmonary arterial hypertension (PAH). Recent studies have shown that cytosolic forkhead box protein O1 (FoxO1) activates autophagy in cancer cells. Paclitaxel has been found to potentially reverse PAH progression. However, the role of FoxO1 and the effects of paclitaxel on autophagy in the pathogenesis of PAH remain unknown. PAH was generated by intraperitoneal injection of monocrotaline (MCT) to rats. The right ventricular systolic pressure (RVSP), the right ventricle hypertrophy index (RV/LV+S), and the percentage of medial wall thickness (%MT) were used to detect the development of PAH. Hematoxylin and eosin staining was performed to measure pulmonary vascular remodeling. The protein level, phosphorylation, and nucleus translocation of FoxO1 and the levels of LC3A, LC3B, and Beclin-1 were examined by immunoblotting. The results showed that in spite of reduced expression of FoxO1, elevated phosphorylation of FoxO1 caused most of FoxO1 accumulating in cytosolic fraction in MCT-PAH rats. Autophagy was also activated in the MCT-PAH group. In cultured rat pulmonary arterial smooth muscle cells (PASMCs), knockdown of FoxO1 markedly blocked autophagy activation, indicating that elevation of cytosolic FoxO1 stimulates autophagy activation. Treatment of PAH rats with paclitaxel reduced FoxO1 phosphorylation and increased FoxO1 nuclear accumulation, despite increased FoxO1 expression, therefore suppressed autophagy, finally reduced elevated RVSP, RV/LV+S, and %MT in MCT-induced PAH. Taken together, paclitaxel inhibits pulmonary vascular remodeling by FoxO1-mediated autophagy suppression, suggesting that paclitaxel might be a novel therapeutic agent for the prevention and treatment of PAH.


Assuntos
Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico , Proteínas do Tecido Nervoso/metabolismo , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Hipertensão Arterial Pulmonar/tratamento farmacológico , Hipertensão Arterial Pulmonar/metabolismo , Animais , Autofagia/efeitos dos fármacos , Células Cultivadas , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Monocrotalina , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/genética , Hipertensão Arterial Pulmonar/induzido quimicamente , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/patologia , Ratos Sprague-Dawley
16.
Pulm Circ ; 9(1): 2045894018816977, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30430898

RESUMO

The upregulation of Sphingosine kinase 1 (SphK1) expression and accompanied sphingosine-1-phosphate (S1P) production have been reported to contribute to the proliferation of pulmonary artery smooth muscle cells (PASMC) and pulmonary arterial remodeling. However, the molecular mechanisms of SphK1/S1P upregulation in PASMC and the specific mechanisms of how SphK1/S1P pathway promotes PASMC proliferation remain largely unclear. This study aims to address these issues. Here, we demonstrated that TGF-ß1 significantly upregulated SphK1 expression and S1P production by promoting the phosphorylation of Smad2/3 in PASMC. Further study indicated that SphK1/S1P pathway mediated TGF-ß1-induced Notch3 activation in PASMC. In addition, we showed that TGF-ß1 significantly induced proliferation of PASMC, while pre-inhibition of Smad2/3 phosphorylation with SB431542 or silencing SphK1 using small interfering RNA in advance, or pre-blocking Notch3 pathway with N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), attenuated TGF-ß1-induced PASMC proliferation. Taken together, our study indicates that Smad2/3/SphK1/S1P/Notch3 pathway mediates TGF-ß1-induced PASMC proliferation and suggests this pathway as a potential therapeutic target in the prevention and treatment of pulmonary hypertension.

17.
J Pharm Pharmacol ; 71(3): 417-428, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30537209

RESUMO

OBJECTIVES: The aim of this study was to evaluate the neuroprotective effects of SalB on high glucose (HG)-induced excessive autophagy and apoptosis in vitro. METHODS: The proliferation and apoptosis of RSC96 cells were determined using the MTT assay and flow cytometry, respectively. Western blot analysis was performed to examine the expression of autophagy and apoptosis-related proteins. RT-PCR and flow cytometry were manipulated to examine the level of Bcl-2. The signals of autophagy markers were detected using immunofluorescence methods. KEY FINDINGS: We found that HG significantly reduced RSC96 cell's proliferation and induced apoptosis. What's more, HG increased the level of autophagy and apoptosis-related proteins. However, these effects were reversed by SalB. In addition, we also found that 3-MA decreased the expression of LC3A/B and Beclin1, while the JNK inhibitor SP600125 reduced the levels of phosphorylated JNK, LC3A/B and Beclin1. CONCLUSIONS: High glucose not only induced apoptosis but also caused autophagic cell death by activating the JNK pathway. These effects prevented by SalB in an opposite manner.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Benzofuranos/farmacologia , Neuropatias Diabéticas/prevenção & controle , Doenças do Sistema Nervoso Periférico/prevenção & controle , Animais , Antracenos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Neuropatias Diabéticas/metabolismo , Glucose/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
18.
Arch Med Sci ; 14(6): 1204-1211, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30393475

RESUMO

INTRODUCTION: Interleukin-17A (IL-17A), a pro-inflammatory cytokine, plays an important role in the pathogenesis of asthma. A number of studies have investigated the relationship between IL-17A rs2275913 polymorphism and risk of asthma. However, the results obtained are inconclusive. The aim of this meta-analysis is to clarify the relationship between IL-17A rs2275913 polymorphism and asthma risk. MATERIAL AND METHODS: Searches were conducted in PubMed, Web of Science, Elsevier, Google Scholar, Wanfang and Chinese National Knowledge Infrastructure (CNKI) databases, and data were extracted from eligible studies by two independent reviewers. The pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were calculated. Publication bias, heterogeneity and sensitivity analysis were also assessed. RESULTS: Ten studies with a total of 5016 subjects were included. Overall, the results indicated a significant association between the IL-17A rs2275913 polymorphism and the risk of asthma (G vs. A: OR = 0.866, 95% CI: 0.789-0.951, p = 0.003; GG+GA vs. AA: OR = 0.752, 95% CI: 0.633-0.895, p = 0.001). In subgroup analysis by age and ethnicity, the G allele of rs2275913 in IL-17A was significantly associated with a reduced risk of asthma in children and Asians. CONCLUSIONS: The results of this meta-analysis indicate that the G allele of rs2275913 in IL-17A is a protective factor for the development of asthma.

19.
Exp Cell Res ; 371(2): 379-388, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30180991

RESUMO

Up-regulation of mammalian COP9 signalosome subunit 6 (CSN6) and consequent reduction of SCF ubiquitin ligase substrate receptor ß-transduction repeat-containing protein (ß-TrCP) have been shown to be associated with cancer cells proliferation. However, it is unclear whether CSN6 and ß-TrCP are also involved in PDGF-induced pulmonary arterial smooth muscle cells (PASMCs) proliferation. This study aims to address this issue and further explore its potential mechanisms. Our results indicated that PDGF phosphorylated Akt, stimulated PASMCs proliferation; while inhibition of PDGF receptor (PDGFR) by imatinib prevented these effects. PDGF further up-regulated CSN6 protein expression, this was accompanied with ß-TrCP reduction and increase of Cdc25A. Inhibition of PDGFR/PI3K/Akt signaling pathway reversed PDGF-induced such changes and cell proliferation. Prior transfection of CSN6 siRNA blocked PDGF-induced ß-TrCP down-regulation, Cdc25A up-regulation and cell proliferation. Furthermore, pre-treatment of cells with MG-132 also abolished PDGF-induced ß-TrCP reduction, Cdc25A elevation and cell proliferation. In addition, pre-depletion of Cdc25A by siRNA transfection suppressed PDGF-induced PASMCs proliferation. Taken together, our study indicates that up-regulation of CSN6 by PDGFR/PI3K/Akt signaling pathway decreases ß-TrCP by increasing its ubiquitinated degradation, and thereby increases the expression of Cdc25A, which promotes PDGF-induced PASMCs proliferation.


Assuntos
Complexo do Signalossomo COP9/genética , Miócitos de Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Animais , Complexo do Signalossomo COP9/antagonistas & inibidores , Complexo do Signalossomo COP9/metabolismo , Proliferação de Células/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Regulação da Expressão Gênica , Mesilato de Imatinib/farmacologia , Leupeptinas/farmacologia , Masculino , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
20.
Life Sci ; 210: 140-149, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30179628

RESUMO

AIMS: This study aims to explore the molecular mechanisms underlying sphingosine kinase 1 (SphK1) inducing pulmonary vascular remodeling and resveratrol suppressing pulmonary arterial hypertension (PAH). MATERIAL AND METHODS: monocrotaline (MCT) was used to induce PAH in rats. The right ventricular systolic pressure (RVSP), right ventricle hypertrophy index (RVHI) and histological analyses including hematoxylin and eosin staining, the percentage of medial wall thickness (%MT), α-SMA staining and Ki67 staining were performed to evaluate the development of PAH. Protein levels of SphK1, nuclear factor-kappaB (NF-κB)-p65 and cyclin D1 were determined using immunoblotting. Sphingosine-1-phosphate (S1P) concentration was measured using enzyme-linked immunosorbent assay. KEY FINDINGS: SphK1 protein level, S1P production, NF-κB activation and cyclin D1 expression were significantly increased in MCT-induced PAH rats. Inhibition of SphK1 by PF543 suppressed S1P synthesis and NF-κB activation and down-regulated cyclin D1 expression in PAH rats. Suppression of NF-κB by pyrrolidine dithiocarbamate (PDTC) also reduced cyclin D1 expression in PAH model. Treatment of PAH rats with either PF543 or PDTC dramatically decreased RVSP, RVHI and %MT and reduced pulmonary arterial smooth muscle cells proliferation and pulmonary vessel muscularization. In addition, resveratrol effectively inhibited the development of PAH by suppression of SphK1/S1P-mediated NF-κB activation and subsequent cyclin D1 expression. SIGNIFICANCE: SphK1/S1P signaling induces the development of PAH by activation of NF-κB and subsequent up-regulation of cyclin D1 expression. Resveratrol inhibits the MCT-induced PAH by targeting on SphK1 and reverses the downstream changes of SphK1, indicating that resveratrol might be a therapeutic agent for the prevention of PAH.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Monocrotalina/toxicidade , NF-kappa B/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Estilbenos/farmacologia , Remodelação Vascular/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Resveratrol , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA