Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 137: 112528, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38908086

RESUMO

Low back pain due to epidural fibrosis is a major complication after spine surgery. Macrophages infiltrate the wound area post laminectomy, but the role of macrophages in epidural fibrosis remains largely elusive. In a mouse model of laminectomy, macrophage depletion decreased epidural fibrosis. CD146, an adhesion molecule involved in cell migration, is expressed by macrophages. CD146-defective macrophages exhibited impaired migration, which was mediated by reduced expression of CCR2 and suppression of the MAPK/ERK signaling pathway. CD146-defective macrophages suppress the MAPK/ERK signaling pathway by increasing Erdr1. In vivo, CD146 deficiency decreased macrophage infiltration and reduced extracellular matrix deposition in wound tissues. Moreover, the anti-CD146 antibody AA98 suppressed macrophage infiltration and epidural fibrosis. Taken together, these findings demonstrated that CD146 deficiency alleviates epidural fibrosis by decreasing the migration of macrophages via the Erdr1/ERK/CCR2 pathway. Blocking CD146 and macrophage infiltration may help alleviate epidural fibrosis.


Assuntos
Antígeno CD146 , Fibrose , Macrófagos , Camundongos Endogâmicos C57BL , Receptores CCR2 , Animais , Receptores CCR2/metabolismo , Receptores CCR2/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Antígeno CD146/metabolismo , Antígeno CD146/genética , Movimento Celular , Camundongos Knockout , Espaço Epidural/patologia , Masculino , Sistema de Sinalização das MAP Quinases/imunologia , Laminectomia , Modelos Animais de Doenças , Transdução de Sinais , Humanos
2.
Altern Ther Health Med ; 30(2): 183-187, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37856813

RESUMO

The purpose of this paper was to construct a prognostic model, miRNA-mRNA regulatory network and protein-protein interaction (PPI) network for lung squamous cell carcinoma (LUSC) used data from the cancer genome atlas (TCGA) database. In this study, we first downloaded and sorted out the expression matrix containing 19962 mRNA transcripts (including 502 LUSC and 51 normal control (NC) samples) and the expression matrix containing 2205 miRNA transcripts (including 478 LUSC and 45 NC samples) from the TCGA database. We obtained 389 differentially expressed miRNAs (DE-miRNAs), of which 305 were upregulated and 84 down-regulated DE-miRNAs. Next, a total of 7 prognosis-related DE-miRNAs (PDE-miRNAs) were identified by Cox regression analysis, and the prognosis model consisting of three PDE-miRNAs (hsa-miR-4746-5p, hsa-miR-556-3p and hsa-miR-489-3p) was optimized. Then, we drew the survival curves and found that the survival rates of the three PDE-miRNA high and low expression groups and the survival rates of the high-risk and low-risk patients in the prognosis model had significant statistical differences. In addition, the receiver operating characteristics (ROC) curve analysis and independent prognostic analysis confirmed that the prognostic model we built has a relatively accurate ability to predict the grouping and prognosis of LUSC patients. Finally, Cox regression analysis were used to construct the miRNA-mRNA regulatory network, which showed the regulatory relationship between PDE-miRNAs and targeted mRNAs. Moreover, we constructed the PPI network composed of 145 targeted mRNAs and the subnetwork composed of 10 hub-targeted mRNAs (FCGR3A, IL13, CCR2, PPARGC1A, FCGR3B, ACSL1, PLXNA4, LPL, KAT2B and AOC3), which showed the interaction between targeted mRNAs. The above results indicated that the prognosis model we built can predict LUSC patients relatively accurately. The miRNA-mRNA regulatory network and the PPI network of targeted mRNAs illustrated the regulatory mechanisms and interactions between RNAs, which were of certain reference significance for us to further understand the molecular pathogenesis of LUSC and for clinical early diagnosis and treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , MicroRNAs , Humanos , Prognóstico , RNA Mensageiro/genética , Redes Reguladoras de Genes , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/diagnóstico , Pulmão
3.
Cancer Biother Radiopharm ; 35(5): 362-370, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32380843

RESUMO

Background: Long noncoding RNA nuclear enriched abundant transcript 1 (NEAT1) has been reported to play a promotive role in nonsmall cell lung cancer (NSCLC) progression through microRNAs (miRNAs). However, the exact influence and mechanism of NEAT1 were unsatisfied. Methods: Quantitative real-time polymerase chain reaction was applied to examine the expression of NEAT1 and miR-153-3p. The cell proliferation ability, apoptosis rate, migration, and invasion were measured by Cell Counting Kit-8 (CCK8) assay, flow cytometry, and transwell assay, respectively. The epithelial-mesenchymal transition process and Wnt/ß-catenin signaling pathway were verified by Western blot. The interaction between NEAT1 and miR-153-3p was confirmed by dual-luciferase reporter assay and RNA immunoprecipitation assay. Results: These data showed that NEAT1 is highly expressed in NSCLC tissues and cell lines. Knockdown of NEAT1 suppresses cell proliferation, invasion, migration, and induces the cell apoptosis in NSCLC cell lines. At the same time, NEAT1 directly interacts with miR-153-3p in NSCLC. In addition, upregulation of miR-153-3p inhibits the cell progression, and miR-153-3p inhibitor recovers the inhibition effect of si-NEAT1 in NSCLC cell lines. Subsequently, si-NEAT1 inhibits Wnt/ß-catenin signaling pathway, which is reactivated by miR-153-3p inhibitor. Conclusions: Knockdown of NEAT1 could suppress cell proliferation, migration, and invasion of NSCLC while promoting cell apoptosis through sponging miR-153-3p.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Pulmão/patologia , Pulmão/cirurgia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Masculino , Pessoa de Meia-Idade , Pneumonectomia , RNA Longo não Codificante/genética , Regulação para Cima , Via de Sinalização Wnt/genética
4.
Oncol Lett ; 18(2): 1548-1556, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31423222

RESUMO

Accumulating evidence suggests that acetyl-CoA acetryltransferase 1 (ACAT-1) may mediate tumor development and metastasis. However, the specific function served by ACAT-1 in lung cancer is not well understood. Therefore, the present study initially verified that ACAT-1 was overexpressed in Lewis lung carcinoma (LLC) tissues compared with non-LLC mice and that this overexpression promoted the proliferation, invasion and metastasis of these LLC samples. Western blotting, immunofluorescence microscopy and flow cytometry allowed the present study to determine that the ACAT-1 inhibitor avasimibe significantly reduced the expression of ACAT-1 in LLC compared with LLC cells that are not treated with avasimibe (P<0.05). A combination of Cell Counting Kit-8 and wound healing assays demonstrated that downregulating ACAT-1 expression sufficiently inhibited the proliferation of LLC cells. Avasimibe promoted LLC cell apoptosis as assessed by a Annexin V/propidium iodide double staining assay. Furthermore, avasimibe inhibited tumor growth in vivo and improved immune responses, with tissue biopsies from LLC model mice exhibiting higher levels of ACAT-1 compared with in healthy controls. Altogether, the results of the present study reveal that avasimibe may inhibit the progression of LLC by downregulating the expression of ACAT-1, which may thus be a potential novel therapeutic target for lung cancer treatment.

5.
Pathol Res Pract ; 215(7): 152422, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31079851

RESUMO

BACKGROUND: Triple-negative breast cancer (TNBC) was known as a fast-growing and an aggressive tumor. Cisplatin is the effective cytotoxic drug used for the treatment of TNBC. In addition, apatinib, a VEGFR2 inhibitor, exhibits antitumor activity in patients with TNBC. However, the effects of combination of apatinib with cisplatin on TNBC remain unclear. Thus, this study aimed to investigate the effects of apatinib in combination with cisplatin on MDA-MB-231 cells. METHODS: Immunohistochemistry was used to detect the expression of VEGFR2. In addition, CCK-8, flow cytometric, transwell assays were used to measure the cell proliferation, apoptosis, migration and invasion, respectively. Moreover, western blotting was used to detect the expressions of Bax, active caspase 3, p-VEGFR2, p-Akt and p-mTOR. RESULTS: VEGFR2 was significantly upreguated in patients with TNBC. In addition, the inhibitory effects of cisplatin on the proliferation, migration and invasion of MDA-MB-231 cells were enhanced by apatinib. Moreover, apatinib increased cisplatin-induced apoptosis on MDA-MB-231 cells via increasing the level of Bax and active caspase 3 and decreasing the expression of Bcl-2. Importantly, apatinib enhanced anti-tumor effect of cisplatin on MDA-MB-231 cells via inhibiting the levels of p-VEGFR2, p-Akt and p-mTOR. CONCLUSION: Our findings indicated that apatinib enhanced the anti-tumor effects of cisplatin on MDA-MB-231 cells via inhibition of VEGFR2. Thus, the combination of apatinib with cisplatin may serve as a potential approach in the treatment of patients with TNBC.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Piridinas/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Quimioterapia Combinada , Feminino , Humanos , Pessoa de Meia-Idade , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Onco Targets Ther ; 11: 883-890, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29497319

RESUMO

Apatinib, a novel small molecule tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor-2, was approved for metastatic gastric adenocarcinoma in China in Oct 2014. This is the first report on its use for advanced colorectal cancer as a kind of third-line therapy to date. Here we report two Chinese patients who presented with metastatic colorectal cancer who received apatinib 850 mg daily as a third-line therapy. Both the patients achieved favorable benefits in outcomes after the administration of apatinib. Patient 1 benefited 4 months progression-free survival and 11 months overall survival, while patient 2's progression-free survival was over 10 months. Both the patients presented hand-foot syndrome, and one of them suffered a slight impairment of liver function, mild elevated blood pressure, and proteinuria. But these adverse events were manageable with symptomatic treatment and dose reduction or a short-time drug withdrawal.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA