Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Chin Med ; 19(1): 40, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38433216

RESUMO

BACKGROUND: Pyroptosis has been demonstrated being closely associated with the inflammatory progression in chronic rhinosinusitis (CRS). However, platycodon D (PLD) has emerged as a key anti-inflammatory mediator in the inflammatory progression of various respiratory diseases. This study aims at investigating whether PLD could reduce inflammatory progression of CRS by inhibiting pyroptosis. METHODS: Nasal mucosal tissues from patients with CRS and the control group (simple nasal septal deviation) were analyzed for morphological difference using hematoxylin & eosin staining and for the expression of pyroptosis-related makers by immunofluorescence (IF). Human nasal epithelial cells (HNEpCs) were cultured and co-stimulated with lipopolysaccharide (LPS)/adenosine triphosphate (ATP) to construct an in vitro cellular model simulating CRS. After pretreatment with PLD, EthD-I staining, TUNEL staining, transmission electron microscopy (TEM), and GSDMD-NT detection were performed to evaluate pyroptosis markers. The NLRP3 inflammasome was detected by IF and western blotting (WB). Reactive oxygen species (ROS) were detected by H2DCFDA staining, and mitochondrial membrane potential was evaluated by JC-1 staining. Mitochondrial morphology and structure were observed using TEM. The Nrf2/HO-1 antioxidant signaling pathway was detected using WB. RESULTS: The nasal mucosa structure of patients with CRS exhibited significant damage, with a marked increase in the expression of pyroptosis-related proteins compared with the control group. LPS/ATP co-stimulation resulted in an increased expression of IL-18 and IL-1ß in HNEpCs, causing significant damage to nuclear and cell membranes, GSDMD-NT accumulation around the cell membrane, and intracellular NLRP3 inflammasome activation. Furthermore, it led to increased ROS expression, significantly decreased mitochondrial membrane potential, and damaged mitochondrial structure. However, pretreatment with PLD significantly reversed the aforementioned trends and activated the Nrf2/HO-1 antioxidant signaling pathway. CONCLUSIONS: The results of this study confirm that NLRP3-mediated pyroptosis plays a crucial role in the pathological process of nasal mucosal impairment in patients with CRS. PLD inhibits NLRP3-mediated pyroptosis, preventing inflammatory damage in HNEpCs of patients with CRS by activating the Nrf2/HO-1 antioxidant signaling pathway, which in turn reduces ROS production and ameliorates mitochondrial damage.

2.
Cell Biol Toxicol ; 40(1): 14, 2024 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-38376606

RESUMO

BACKGROUND: RING Finger Protein 115 (RNF115), a notable E3 ligase, is known to modulate tumorigenesis and metastasis. In our investigation, we endeavor to unravel the putative function and inherent mechanism through which RNF115 influences the evolution of thyroid carcinoma (THCA). METHODS: We analyzed RNF115 expression in THCA using the Cancer Genome Atlas (TCGA) database. The influence of RNF115 on the progression of THCA was evaluated using both in vitro and in vivo experimental approaches. The protein regulated by RNF115 was identified through bioinformatics analysis, and its biological significance was further explored. RESULTS: In both THCA tissues and cells, RNF115 showed elevated expression levels. Enhanced expression of RNF115 fostered cell proliferation, tumor growth, and the exacerbation of epithelial-mesenchymal transition (EMT) in THCA, while also promoting tumor lung metastasis. Bioinformatics analysis identified cyclin-dependent kinase 10 (CDK10) as a downstream target of RNF115, which was found to be ubiquitinated and degraded by RNF115 in THCA cells. Functionally, overexpression of CDK10 was found to counteract the promotion of malignant phenotype in THCA induced by RNF115. From a mechanistic perspective, RNF115 activated the Raf-1 pathway and enhanced cancer cell cycle progression by degrading CDK10 in THCA cells. CONCLUSION: RNF115 triggers cell proliferation, EMT, and tumor metastasis by ubiquitinating and degrading CDK10. The regulation of the Raf-1 pathway and cell cycle progression in THCA may be profoundly influenced by this process.


Assuntos
Neoplasias Pulmonares , Neoplasias da Glândula Tireoide , Ubiquitina-Proteína Ligases , Humanos , Carcinogênese/genética , Transformação Celular Neoplásica , Quinases Ciclina-Dependentes , Neoplasias da Glândula Tireoide/genética , Ubiquitina-Proteína Ligases/genética
3.
Pathol Res Pract ; 243: 154360, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36801505

RESUMO

TRG-AS1 has been proved to inhibit cancer progression, whereas its effect on bone metastases of breast cancer is unknown. In this study, we determined breast cancer patients with disease free survival is longer in breast cancer patients with high TRG-AS1 expression. Moreover, TRG-AS1 was downregulated in breast cancer tissues and even lower in bone metastatic tumor tissues. Compared with parental breast cancer cell MDA-MB-231, TRG-AS1 expression was downregulated in MDA-MB-231-BO cells with strong bone-metastatic characteristics. Next, the binding sites of miR-877-5p on TRG-AS1 and WISP2 mRNA were predicted and result showed that miR-877-5p could bind to 3'UTR of TRG-AS1 and WISP2. Subsequently, BMMs and MC3T3-E1 cells were cultured in the conditioned media of MDA-MB-231 BO cells transfected with TRG-AS1 overexpression vector, shRNA and/or miR-877-5p mimics or inhibitor and/or overexpression vector and small interfering RNA of WISP2. TRG-AS1 silencing or miR-877-5p overexpression promoted MDA-MB-231 BO cell proliferation and invasion. TRG-AS1 overexpressing reduced TRAP positive cells, decreased TRAP, Cathepsin K, c-Fos, NFATc1 and AREG expression in BMMs, and promoted OPG, Runx2 and Bglap2 expression, and decreased RANKL expression in MC3T3-E1 cells. Silencing WISP2 rescued the effect of TRG-AS1 on BMMs and MC3T3-E1 cells. In vivo results showed that tumor volumes significantly decreased in mice injected with LV-TRG-AS1 transfected MDA-MB-231 cells. TRG-AS1 knockdown markedly reduced the number of TRAP+ cells and the percentage of Ki-67+ cells and decreased E-cadherin expression in xenograft tumor mice. In summary, TRG-AS1 acts an endogenous RNA, inhibited breast cancer bone metastasis by competitively binding with miR-877-5p to upregulate WISP2 expression.


Assuntos
Neoplasias da Mama , MicroRNAs , RNA Longo não Codificante , Animais , Feminino , Humanos , Camundongos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno/genética
4.
Front Oncol ; 12: 1010131, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591522

RESUMO

The nasal mucosa, which performs the crucial functions of filtering, humidifying and temperature regulation, is one of the most vulnerable areas of nasopharyngeal carcinoma (NPC) patients after radiotherapy (RT). Following RT, NPC patients experience a series of pathological changes in the nasal mucosa, ultimately leading to physiological dysfunction of the nasal epithelium. This article systematically reviews the clinical and pathological manifestations of RT-related nasal damage in NPC patients and summarizes the potential mechanism of damage to the human nasal epithelium by RT. Finally, we outline the current mechanistic models of nasal epithelial alterations after RT in NPC patients and provide additional information to extend the in-depth study on the impairment mechanisms of the nasal mucosa resulting from RT. We also describe the relationship between structural and functional alterations in the nasal mucosa after RT to help mitigate and treat this damage and provide insights informing future clinical and fundamental investigations.

5.
Mol Med Rep ; 14(4): 3421-8, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27511210

RESUMO

Gastric carcinoma (GC) ranks as the second most common cause of cancer­associated mortality worldwide. Emerging evidence has suggested a potential novel therapeutic strategy based on the ability of cancer stem cells (CSCs) to trigger tumorigenesis. MicroRNAs (miRNAs) have previously been implicated in CSC formation and regulation of their functional characteristics. In the current study, a significant upregulation of miR­483­5p levels was demonstrated in spheroid body­forming cells (P<0.01) by reverse transcription­quantitative polymerase chain reaction, which were isolated from the MKN­45 gastric cancer cell line and possessed gastric CSC (GCSC) properties. An MTT assay demonstrated that overexpression of miR­483­5p by transfection with miR­483­5p mimics significantly increased cell proliferation and Annexin V­propidium iodide staining indicated the suppression of cell apoptosis, suggesting that miR­483­5p has an important function in GCSC growth. Notably, Transwell and sphere formation assays demonstrated that miR­483­5p elevation promoted GCSC invasion and cell self­renewal ability, respectively. Further western blotting assays demonstrated that miR­483­5p upregulation induced an increase in the protein expression levels of ß­catenin and its downstream target molecules, including cyclin D1, Bcl­2 and matrix metalloproteinase 2, indicating that miR­483­5p activates Wnt/ß­catenin signaling. Inhibition of this pathway by ß­catenin small interfering RNA transfection attenuated the miR­483­5p­induced effects on cell growth, invasion and self­renewal. These results demonstrate that miR­483­5p may act as an oncogene to promote the development of GC by regulating GCSC growth, invasion and self­renewal via the Wnt/ß­catenin signaling pathway. Thus, the present study suggests that miR­483­5p may be a promising therapeutic target against GC.


Assuntos
MicroRNAs/genética , Invasividade Neoplásica/genética , Células-Tronco Neoplásicas/patologia , Neoplasias Gástricas/genética , Estômago/patologia , Regulação para Cima , Via de Sinalização Wnt , Linhagem Celular Tumoral , Proliferação de Células , Mucosa Gástrica/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , beta Catenina/metabolismo
6.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 31(2): 154-7, 2013 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-23662556

RESUMO

OBJECTIVE: To investigate the effect of modified bone morphogenetic protein-2 polylactic acid nanospheres (BMP-2-PLA-Ns) sustained-release system on rabbit mandibular defect repair. METHODS: The polylactic acid s nanospheres (PLA-Ns) and BMP-2-PLA-Ns were prepared by ultrasonic emulsification after graft polymerization. Forty-five rabbits were randomly divided into 3 groups: Blank group, PLA-Ns gel group(control group), and BMP-2-PLA-Ns gel group (experimental group). The rabbit mandibular defect models were established. The defect area of control group was implanted with PLA-Ns gel, meanwhile, the experimental group was implanted with BMP-2-PLA-Ns gel, the blank group experienced no special handling. Rabbits were killed in 1, 2, 4 weeks after operation and the iconography, hematine eosin(HE) staining and PCNA immunohistochemistry were used to detect the reparative effect on rabbit mandible defects. RESULTS: Image observation showed that bone defect repair in the experimental group was well and the shadow was not obvious. Better repair effect was seen compared with the control group and blank group. HE staining showed that the experimental group and the control group had a large number of neovascularization and secondary callus formation, callus in experimental group was obviously higher than that of control group and blank group. Immunohistochemical observation showed that the experimental group's PCNA positive chondrocytes were more than those in the control group and the blank group in the first 2 weeks; all groups of PCNA positive cells were rare in the fourth week, PCNA positive expression rate of the fourth week was lower than that of the first 2 weeks. CONCLUSION: The modified BMP-2-PLA-Ns sustained-release system promotes mandibular defect repair obviously.


Assuntos
Proteína Morfogenética Óssea 2 , Nanosferas , Animais , Preparações de Ação Retardada , Ácido Láctico , Mandíbula , Poliésteres , Polímeros , Coelhos , Procedimentos de Cirurgia Plástica
7.
FEBS Lett ; 586(19): 3508-21, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22958893

RESUMO

MicroRNAs (miRNAs) represent a class of small non-coding RNAs that regulate gene expression at the post-transcriptional levels. Recent studies show that miRNAs may function as oncogenes or tumor suppressor genes. In this study, we demonstrated that miR-518b was down-regulated in esophageal squamous cell carcinoma (ESCC) tissues and correlated with metastasis and survival. miR-518b suppressed the proliferation by inducing apoptosis and repressed the invasion in ESCC cells, but had no effect on the cell cycle. Furthermore, Rap1b was revealed to be directly regulated by miR-518b. These findings indicate that miR-518b may function as a tumor suppressor by targeting Rap1b in the development of ESCC and has important clinical and prognostic value.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , MicroRNAs/genética , Proteínas rap de Ligação ao GTP/metabolismo , Idoso , Apoptose/genética , Sequência de Bases , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundário , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Intervalo Livre de Doença , Regulação para Baixo , Neoplasias Esofágicas/metabolismo , Feminino , Humanos , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteínas rap de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA