Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Analyst ; 149(17): 4378-4387, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-38995156

RESUMO

Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer and a major cause of cancer-related mortality worldwide. Small extracellular vesicles (sEVs) are heterogeneous populations of membrane-structured vesicles that can be found in many biological fluids and are currently considered as a potential source of disease-associated biomarkers for diagnosis. The purpose of this study was to define the proteomic and phosphoproteomic landscape of urinary sEVs in patients with HCC. Mass spectrometry-based methods were used to detect the global proteome and phosphoproteome profiles of sEVs isolated by differential ultracentrifugation. Label-free quantitation analysis showed that 348 differentially expressed proteins (DEPs) and 548 differentially expressed phosphoproteins (DEPPs) were identified in the HCC group. Among them, multiple phosphoproteins related to HCC, including HSP90AA1, IQGAP1, MTOR, and PRKCA, were shown to be upregulated in the HCC group. Pathway enrichment analysis indicated that the upregulated DEPPs participate in the regulation of autophagy, proteoglycans in cancer, and the MAPK/mTOR/Rap1 signaling pathway. Furthermore, kinase-substrate enrichment analysis revealed activation of MTOR, AKT1, MAP2Ks, and MAPKs family kinases in HCC-derived sEVs, indicating that dysregulation of the MAPK and mTOR signaling pathways may be the primary sEV-mediated molecular mechanisms involved in the development and progression of HCC. This study demonstrated that urinary sEVs are enriched in proteomic and phosphoproteomic signatures that could be further explored for their potential use in early HCC diagnostic and therapeutic applications.


Assuntos
Carcinoma Hepatocelular , Vesículas Extracelulares , Neoplasias Hepáticas , Fosfoproteínas , Proteômica , Carcinoma Hepatocelular/urina , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/urina , Neoplasias Hepáticas/metabolismo , Humanos , Fosfoproteínas/metabolismo , Fosfoproteínas/urina , Vesículas Extracelulares/metabolismo , Proteômica/métodos , Masculino , Serina-Treonina Quinases TOR/metabolismo , Biomarcadores Tumorais/urina , Pessoa de Meia-Idade , Feminino , Proteínas Ativadoras de ras GTPase/metabolismo , Proteoma , Proteína Quinase C-alfa
2.
Anal Chim Acta ; 1301: 342450, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38553121

RESUMO

Molecular imprinting polymers (MIPs) are synthetic receptors as biomimetic materials for various applications ranging from sensing to separation and catalysis. However, currently existing MIPs are stuck to some of the issues including the longer preparation steps and poor performance. In this report, a facile and one-pot strategy by integrating the in-situ growth of magnetic nanoparticles and reversed phase microemulsion oriented molecularly imprinting strategy to develop magnetic molecular imprinted nanocomposites was proposed. Through self-assembling of the template, it brought up highly ordered and uniform arrangement of the imprinting structure, which offered faster adsorption kinetic as adsorption equilibrium was achived within 15 min, higher adsorption capacity (Qmax = 48.78 ± 1.54 µmol/g) and high affinity (Kd = 127.63 ± 9.66 µM) toward paradigm molecule-adenosine monophosphate (AMP) compared to the conventional bulk imprinting. The developed MIPs offered better affinity and superior specificity which allowed the specific enrichment toward targeted phosphorylated peptides from complex samples containing 100-fold more abundant interfering peptides. Interestingly, different types of MIPs can be developed which could targetly enrich the specific phosphorylated peptides for mass spectrometry analysis by simply switching the templates, and this strategy also successfully achieved imprinting of macromolecular peptides. Collectively, the approach showed broad applicability to target specific enrichment from metabolites to phosphorylated peptides and providing an alternative choice for selective recognition and analysis from complex biological systems.


Assuntos
Impressão Molecular , Polímeros , Polímeros/química , Peptídeos , Substâncias Macromoleculares , Adsorção , Impressão Molecular/métodos
3.
Gen Hosp Psychiatry ; 84: 47-59, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37385139

RESUMO

PURPOSE: This meta-analysis was to assess the efficacy of digital psychological interventions to improve physical symptoms (i.e., fatigue, pain, disturbed sleep, and physical well-being) among cancer patients, as well as to evaluate the variables that possibly moderate intervention effects. METHODS: Nine databases were searched for the literature up to February 2023. Two reviewers independently conducted a quality assessment. Effect sizes were reported as the standardized mean difference (Hedge's g) and estimated using a random-effects model. RESULTS: The meta-analysis included 44 randomized clinical trials comprising 7200 adults with cancer. Digital psychological interventions were associated with significant improvements in short-term fatigue (g = -0.33; 95% CI, -0.58 to -0.07) and disturbed sleep (g = -0.36; 95% CI, -0.57 to -0.15), but with non-significant changes in pain (g = -0.23; 95% CI, -0.68 to 0.21) and physical well-being (g = 0.31; 95% CI, -0.18 to 0.80). Additionally, no alleviation in long-term physical symptoms was observed. In subgroup analysis, results suggest that the country significantly moderated the effectiveness of digital psychological interventions in alleviating fatigue. CONCLUSIONS: Digital psychological interventions can be effective for improving short-term fatigue and disturbed sleep in patients with cancer. Clinicians could consider digital psychological interventions as a possible and efficient addition to better manage some of the physical symptoms during and after cancer treatment.


Assuntos
Neoplasias , Intervenção Psicossocial , Adulto , Humanos , Ansiedade/terapia , Neoplasias/complicações , Dor , Fadiga/etiologia , Fadiga/terapia
4.
J Chemother ; 33(7): 476-485, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33825671

RESUMO

This study aims to investigate the effects of C1q-like 1 (C1QL1) on the growth and migration of lung adenocarcinoma (LUAD) cells and the underlying mechanism. The expression of C1QL1 in LUAD tissues and its prognostic value were analyzed using the data from The Cancer Genome Atlas (TCGA) database. To investigate the function of C1QL1, loss-of-function and gain-of-function assays were conducted in Calu-3 cells and LTEP-a-2 cells, respectively. Cell growth was evaluated by CCK-8 and colony formation assays. Transwell assays were performed to assess cell invasive and migratory abilities. qRT-PCR and Western blotting were performed to detect RNA and protein expression, respectively. Firstly, we found that C1QL1 was highly expressed and predicted poor outcomes in LUAD patients from TCGA database. Moreover, the mRNA and protein expression levels of C1QL1 were higher in LUAD cells than that in normal lung cells. Results of functional experiments illustrated that depletion of C1QL1 restrained the growth, invasion and migration of Calu-3 cells, meanwhile over-expression of C1QL1 presented the opposite results in LTEP-a-2 cells. Furthermore, we discovered that down-regulation of C1QL1 elevated the protein level of E-cadherin and reduced the protein levels of N-cadherin, Vimentin and Snail in Calu-3 cells, whereas over-expression of C1QL1 led to the opposite outcomes in LTEP-a-2 cells. Our data indicated that C1QL1 functioned as a crucial driver in LUAD cell growth and motility, which might be achieved by modulating epithelial-mesenchymal transition (EMT). These consequences are of important relevance for the design of therapeutic strategies for LUAD.


Assuntos
Adenocarcinoma de Pulmão/patologia , Complemento C1q/biossíntese , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Biologia Computacional , Humanos , Invasividade Neoplásica/fisiopatologia , Prognóstico , Regulação para Cima
5.
Cell Biochem Biophys ; 78(4): 511-520, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32705535

RESUMO

Esophageal cancer (EC) is a common digestive tract malignant tumor and the clinical outcome of patients with EC after surgery remains unsatisfactory. Hence, it is necessary to identify some effective drugs or methods to improve the prognosis of patients with EC. In this study, we attempted to analyze the potential role of hydroxysafflor yellow A (HSYA) in EC. Combined with The Cancer Genome Atlas (TCGA) and Comparative Toxicogenomics Database (CTD) as well as Database for Annotation, Visualization, and Integrated Discovery (DAVID) website, we tried to identify the related genes and pathways of HSYA. Then we estimated the actions of HSYA on proliferation, invasion and migration, and apoptosis of EC cells using cell counting kit 8, transwell and flow cytometry assays, respectively. At last, the expression of inflammatory protein and signaling pathway-related protein were measured using western blot analysis. Relative protein expression of intercellular adhesion molecule 1 (ICAM1), matrix metallopeptidase 9 (MMP9), tumor necrosis factor (TNF), and vascular cell adhesion molecule 1 (VCAM1) were all upregulated in EC tissues compared with normal tissues and they might be the target gene of HSYA according to bioinformatics analysis. HSYA exerted an inhibitory actions on cells proliferation, invasion, and migration but could accelerate the apoptosis of cells in EC. Moreover, HSYA could inhibit the expression of ICAM1, MMP9, TNF-α, and VCAM1 and induced the expression of phosphor-nuclear transcription factor kappa B p65 (p-P65) and phosphor-I kappa B-alpha (p-IκBα), but it did not influence the expression of P65 and IκBα. HSYA suppressed proliferation, invasion, and migration, simultaneously induce apoptosis of EC cells partly via regulating NF-κB signaling pathway.


Assuntos
Antineoplásicos/farmacologia , Carcinogênese/efeitos dos fármacos , Carthamus tinctorius/química , Chalcona/análogos & derivados , Neoplasias Esofágicas/patologia , NF-kappa B/metabolismo , Quinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chalcona/farmacologia , Descoberta de Drogas , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
Arch Med Sci ; 15(4): 968-978, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31360191

RESUMO

INTRODUCTION: Our study was conducted to prove that miR-509-5p improved the proliferative and invasive abilities of papillary thyroid carcinoma (PTC) cells through inhibiting SFRP1 expression. MATERIAL AND METHODS: QRT-PCR was conducted in order to detect the miR-509-5p expression levels in PTC and normal tissues. The miR-509-5p and SFRP1 mRNA expression levels in PTC cell lines K1, TPC-1, BCPAP and the human normal thyroid cell line HT-ori3 were also detected by qRT-PCR. The transfection was performed using Lipofectamine and lentiviral vectors. Pgcsil-008 was used as the SFRP1 gene vector. Western blot and dual luciferase reporter gene assay were conducted to investigate miR-509-5p's direct regulation on SFRP1. MTT, clone formation, and Transwell assays were adopted to investigate the biological behaviors of PTC cells. TCF/LEF luciferase assays were used to prove that miR-509-5p influenced the Wnt/ß-catenin signaling pathway by regulating SFRP1. RESULTS: MiR-509-5p was overexpressed in PTC cells and tissues in which SFRP1 was down-regulated. MiR-509-5p bound to the 3'-UTR of SFRP1 and therefore partially weakened the proliferative, migrating and invasive activities of PTC cells. MiR-509-5p promoted activation of the Wnt/ß-catenin signaling pathway through down-regulating SFRP1. CONCLUSIONS: MiR-509-5p improved the proliferative, migrating and invasive abilities of PTC cells through inhibiting SFRP1 expression.

7.
Appl Immunohistochem Mol Morphol ; 27(5): 364-372, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29489510

RESUMO

Variant pulmonary adenocarcinoma with intestinal-type molecules shares similar molecular expression with colorectal carcinoma. However, expression of such molecules and their association with survival time with clinicopathologic parameters, such as epidermal growth factor receptor (EGFR) gene status in other types of pulmonary adenocarcinoma, has been rarely demonstrated. Sixty patients with resected pulmonary adenocarcinoma were divided into the enteric differentiation group (I group, n=30) and the usual group (U group, n=30). Immunohistochemical staining was used to assess the expression of carcinoembryonic antigen (CEA), Villin, CK20, and caudal-related homeobox 2 (CDX2). EGFR gene status was examined by Fluorescence Quantitative polymerase chain reaction. Kaplan-Meier survival curve was drawn by GraphPad Prism 5.0 software. The results showed that there was a significant difference between the 2 groups (P<0.05) in the expression of Villin, CK20, and CDX2, whereas the expression of CEA showed no significant difference (P>0.05). Compared with the U group, patients in the I group were mainly female individuals and in clinical stages III to IV, prone to lymph node metastasis (P<0.05). The patients in the I group with CDX2CK20 phenotype (tumor size>5 cm) had a shorter survival time (P<0.05), and EGFR gene status was not associated with median survival time and the expression of CEA, Villin, CK20, and CDX2 (P>0.05). Thus, our research indicates that patients with enteric differentiation have unique clinical characteristics and different prognosis, which may play important roles in diagnosis and choosing therapeutic strategies for pulmonary adenocarcinoma patients in clinical practice.

8.
Thorac Cancer ; 9(1): 105-111, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29130641

RESUMO

BACKGROUND: Lung cancer is one of the most frequently diagnosed malignancies in the world, thus developing novel anticancer reagents for lung cancer treatment is critical. METHODS: We performed cell counting kit-8 and cell colony formation assays to investigate the role of UNBS5162 in the proliferation of A549 cells. Invasion and migration assays were applied to study the inhibitory effect of UNBS5162 on non-small cell lung cancer cells. To detect the effect of UNBS5162 on A549 cell apoptosis, Annexin-V fluorescein isothiocyanate and propidium iodide staining methods were used. Protein expression was analyzed using Western blot assay. RESULTS: UNBS5162 not only inhibited proliferation but also decreased invasion and migration in A549 cells. Most cells were intact (96.93%) under control conditions, but the number of intact cells decreased (84.8%) after 24 hours of treatment with UNBS5162, and the number of early and late apoptotic cells significantly increased (P < 0.05). Anti-apoptotic protein Bcl-2 expression in the UNBS5162 group was significantly decreased (P < 0.05), and expression of proapoptotic proteins Bim, Bax, and active caspase-3 were significantly increased (P < 0.05) compared to the control. In the PI3K signaling pathway, phospo-AKT and phospo-mTOR levels were significantly decreased (P < 0.05), while S6K and Cyclin D1 protein levels were significantly decreased in UNBS5162 treated A549 cells (P < 0.05). CONCLUSION: These findings suggest that UNBS5162 could inhibit A549 cell proliferation and metastasis by inhibiting PI3K pathway mediated apoptosis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , Células A549 , Apoptose , Proliferação de Células , Humanos , Naftalimidas , Ureia/análogos & derivados
9.
Mol Med Rep ; 15(5): 2620-2624, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28447753

RESUMO

The present study demonstrated the effect of fucoidan, isolated from Fucus vesiculosus, on cell growth and apoptosis in anaplastic thyroid cancer cells. The cell viability was analyzed using a Cell Counting Kit­8 cell proliferation kit. Diamidino-2-phenylindole and terminal deoxynucleotidyl transferase-mediated dUTP nick­end labeling assays were used to examine the apoptotic effect of fucoidan, which revealed the presence of apoptotic bodies and DNA fragmentation. Fucoidan inhibited the growth of FTC133 and TPC1 ATC cells in a dose­dependent manner. It also induced the apoptosis of FTC133 cells by promoting the expression levels of cleaved poly ADP­ribose polymerase and caspase­3. Significant decreases in the levels expression of hypoxia-inducible factor 1α and vascular endothelial growth factor were observed in the FTC133 cells following treatment of the cells with fucoidan. In addition, inhibition in tube formation and the migration of FTC133 cells were observed in the cells treated with fucoidan, compared with the cells in the control group. Therefore, fucoidan inhibited cell growth, induced apoptosis and suppressed angiogenesis in the thyroid cancer cells.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Neovascularização Patológica , Polissacarídeos/farmacologia , Neoplasias da Glândula Tireoide , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias da Glândula Tireoide/irrigação sanguínea , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia
10.
J Cancer Res Ther ; 10(4): 1013-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25579546

RESUMO

BACKGROUND: Breast cancer is one of the most frequently diagnosed cancers in women. Though death from this disease is mainly caused by the metastases of the aggressive cancer cells, few studies have expounded the aggressive behavior of breast cancer. MATERIALS AND METHODS: We downloaded the gene expression profiles of GSE40057, including four aggressive and six less-aggressive breast cancer cell lines, from Gene Expression Omnibus and identified the differentially expressed genes (DEGs) between the aggressive and less-aggressive samples. An integrated gene regulatory network was built including DEGs, microRNAs (miRNAs), and transcription factors. Then, motifs and modules of the network were identified. Modules were further analyzed at a functional level using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway to study the aggressive behavior of breast cancer. RESULTS: A total of 764 DEGs were found and two modules were filtered from the integrated gene regulatory network. Totally two motifs and modules for DEGs were identified. Significant GO terms associated with cell proliferation and hormone stimulus of the modules were found and the target genes identified were  CAV1, CD44, and TGFßR2. The KEGG pathway analysis discovered that CAV1 and FN1 were significantly enriched in focal adhesion, extracellular matrix (ECM)-receptor interaction, and pathways in cancer. CONCLUSION: Aggressive behavior of breast cancer was proved to be related to cell proliferation and hormone stimulus. Genes such as CAV1, CD44, TGFßR2, and FN1 might be potential targets to diagnose the aggressive behavior of breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Biologia Computacional , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Hormônios/metabolismo , Humanos , MicroRNAs/metabolismo , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA