Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 135: 112283, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38772299

RESUMO

Toll-like receptors (TLRs) play a crucial role in mediating immune responses by recognizing pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), as well as facilitating apoptotic cell (ACs) clearance (efferocytosis), thus contributing significantly to maintaining homeostasis and promoting tissue resolution. In this study, we investigate the impact of TLR agonists on macrophage efferocytosis. Our findings demonstrate that pretreatment with the TLR agonist lipopolysaccharide (LPS) significantly enhances macrophage phagocytic ability, thereby promoting efferocytosis both in vitro and in vivo. Moreover, LPS pretreatment confers tissue protection against damage by augmenting macrophage efferocytic capacity in murine models. Further examination reveals that LPS modulates efferocytosis by upregulating the expression of Tim4.These results underscore the pivotal role of TLR agonists in regulating the efferocytosis process and suggest potential therapeutic avenues for addressing inflammatory diseases. Overall, our study highlights the intricate interplay between LPS pretreatment and efferocytosis in maintaining tissue homeostasis and resolving inflammation.


Assuntos
Lipopolissacarídeos , Macrófagos , Camundongos Endogâmicos C57BL , Fagocitose , Animais , Fagocitose/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Receptores Toll-Like/metabolismo , Receptores Toll-Like/agonistas , Inflamação/tratamento farmacológico , Inflamação/imunologia , Masculino , Humanos , Apoptose/efeitos dos fármacos , Células RAW 264.7 , Proteínas de Membrana/metabolismo , Eferocitose
2.
Technol Cancer Res Treat ; 22: 15330338231161141, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36960492

RESUMO

Introduction: Nuclear respiratory factor 1 (NRF1) is an important regulator involved in mitochondrial biogenesis and energy metabolism. However, the specific mechanism of NRF1 in anoikis and epithelial-mesenchymal transition (EMT) remains unclear. Methods: We examined the effect of NRF1 on mitochondria and identified the specific mechanism through transcriptome sequencing, and explored the relationships among NRF1, anoikis, and EMT. Results: We found that upregulated NRF1 expression led to increased mitochondrial oxidative phosphorylation (OXPHOS) and ATP generation. Simultaneously, a significant amount of ROS is generated during OXPHOS. Alternatively, NRF1 upregulates the expression of ROS-scavenging enzymes, allowing tumor cells to maintain low ROS levels and promoting anoikis resistance and EMT. We also found that exogenous ROS was maintained at a low level by NRF1 in breast cancer cells. Conclusion: our study provides mechanistic insight into the function of NRF1 in breast cancer, indicating that NRF1 may serve as a therapeutic target for breast cancer treatment.


Assuntos
Anoikis , Neoplasias da Mama , Transição Epitelial-Mesenquimal , Fator 1 Nuclear Respiratório , Humanos , Feminino , Linhagem Celular Tumoral , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/genética , Fator 1 Nuclear Respiratório/genética , Fator 1 Nuclear Respiratório/metabolismo , Fosforilação Oxidativa , Homeostase , Anoikis/genética , Trifosfato de Adenosina/biossíntese , Mitocôndrias/metabolismo , Potencial da Membrana Mitocondrial , Espécies Reativas de Oxigênio/metabolismo
3.
BMC Neurosci ; 24(1): 10, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36721107

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common malignant intracranial tumor with a low survival rate. However, only few drugs responsible for GBM therpies, hence new drug development for it is highly required. The natural product Cudraflavone B (CUB) has been reported to potentially kill a variety of tumor cells. Currently, its anit-cancer effect on GBM still remains unknown. Herein, we investigated whether CUB could affect the proliferation and apoptosis of GBM cells to show anti-GBM potential. RESULTS: CUB selectively inhibited cell viability and induced cell apoptosis by activating the endoplasmic reticulum stress (ER stress) related pathway, as well as harnessing the autophagy-related PI3K/mTOR/LC3B signaling pathway. Typical morphological changes of autophagy were also observed in CUB treated cells by microscope and scanning electron microscope (SEM) examination. 4-Phenylbutyric acid (4-PBA), an ER stress inhibitor, restored the CUB-caused alteration in signaling pathway and morphological change. CONCLUSIONS: Our finding suggests that CUB impaired cell growth and induced cell apoptosis of glioblastoma through ER stress and autophagy-related signaling pathways, and it might be an attractive drug for treatment of GBM.


Assuntos
Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Autofagia , Apoptose , Estresse do Retículo Endoplasmático
4.
Free Radic Biol Med ; 194: 1-11, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36436726

RESUMO

Glioblastoma is the most lethal intracranial malignant tumor, for which the five-year overall survival rate is approximately 5%. Here we explored the therapeutic combination of vitamin C and plasma-conditioned medium on glioblastoma cells in culture and as subcutaneous or intracranial xenografts in mice. The combination treatment reduced cell viability and proliferation while promoting apoptosis, and the effects were significantly stronger than with either treatment on its own. Similar results were obtained in the two xenograft models. Vitamin C appeared to upregulate aquaporin-3 and enhance the uptake of extracellular H2O2, while the combination treatment increased intracellular levels of reactive oxygen species including H2O2 and activated the JNK signaling pathway. The cytotoxic effects of the combination treatment were partially reversed by the specific JNK signaling inhibitor SP600125. Our results suggest that the combination of vitamin C and plasma-conditioned medium has therapeutic potential against glioblastoma, and they provide mechanistic insights that may help investigate this and other potential therapies in greater depth.


Assuntos
Antineoplásicos , Glioblastoma , Humanos , Animais , Camundongos , Glioblastoma/metabolismo , Peróxido de Hidrogênio/metabolismo , Meios de Cultivo Condicionados/farmacologia , Ácido Ascórbico/farmacologia , Linhagem Celular Tumoral , Apoptose , Antineoplásicos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Vitaminas/farmacologia
5.
Front Biosci (Landmark Ed) ; 28(12): 341, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38179762

RESUMO

BACKGROUND: Vitexicarpin (VIT), an isoflavone derived from various medicinal herbs, has shown promising anti-tumor activities against multiple cancer cells. However, the understanding of the mechanisms and potential targets of VIT in treating triple-negative breast cancer (TNBC) remains limited. METHODS: The potential VIT targets were searched for in the Super-PRED online database, while the TNBC targets were acquired in the DisGeNET database, and the Veeny database was used to identify the VIT and TNBC targets that overlapped. Then, GO and KEGG enrichment analyses were carried out in the DAVID database. The protein-protein interaction (PPI) network was constructed to acquire the hub targets in the STRING database, and the overall survival analysis of the hub targets was examined in the Kaplan-Meier plotter database. Afterward, molecular docking was performed to evaluate the binding capabilities between VIT and the hub targets. In order to measure the effect of VIT on proliferation, apoptosis, and cell cycle arrest in the TNBC cell lines-MDA-MB-231 and HCC-1937-the Cell Counting Kit-8 (CCK-8) assay and flow cytometry analysis were performed. The Western blot and pull-down assays were used to verify the molecular mechanisms by modulating the hub targets. RESULTS: The network pharmacology results identified a total of 37 overlapping genes that were shared by VIT and TNBC. The results of the PPI network and molecular docking analyses showed that HSP90AA1, CREBBP, and HIF-1A were key targets of VIT against TNBC. However, the pull-down results suggested that VIT could directly bind to HSP90AA1 and HIF-1A, yet not to CREBBP. The results of the in vitro tests showed that VIT decreased proliferation and induced apoptosis in MDA-MB-231 and HCC-1937 cells, in a dose-dependent manner, while the cell cycle arrest occurred at the G2 phase. Mechanistically, the Western blot assay demonstrated that VIT decreased the expression of HSP90AA1, CREBBP, and HIF-1A. CONCLUSIONS: VIT inhibited growth and induced apoptosis of TNBC cells by modulating HIF-1A, HSP90AA1, and CREBBP expression. Our findings suggest that VIT is a potential drug for TNBC therapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Simulação de Acoplamento Molecular , Farmacologia em Rede
6.
Front Oncol ; 12: 917537, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091118

RESUMO

Ferroptosis is one of the programmed modes of cell death that has attracted widespread attention recently and is capable of influencing the developmental course and prognosis of many tumors. Glioma is one of the most common primary tumors of the central nervous system, but effective treatment options are very limited. Ferroptosis plays a critical role in the glioma progression, affecting tumor cell proliferation, angiogenesis, tumor necrosis, and shaping the immune-resistant tumor microenvironment. Inducing ferroptosis has emerged as an attractive strategy for glioma. In this paper, we review ferroptosis-related researches on glioma progression and treatment.

7.
J Clin Med ; 11(13)2022 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-35806945

RESUMO

Glioblastoma multiforme (GBM) is an aggressive brain tumor with high risks of recurrence and mortality. Chemoradiotherapy resistance has been considered a major factor contributing to the extremely poor prognosis of GBM patients. Therefore, there is an urgent need to develop highly effective therapeutic agents. Here, we demonstrate the anti-tumor effect of morusin, a typical prenylated flavonoid, in GBM through in vivo and in vitro models. Morusin showed selective cytotoxicity toward GBM cell lines without harming normal human astrocytes when the concentration was less than 20 µM. Morusin treatment significantly induced apoptosis of GBM cells, accompanied by the activation of endoplasmic reticulum (ER) stress, and the appearance of cytoplasmic vacuolation and autophagosomes in cells. Then, we found the ER stress activation and cytotoxicity of morusin were rescued by ER stress inhibitor 4-PBA. Furthermore, morusin arrested cell cycle at the G1 phase and inhibited cell proliferation of GBM cells through the Akt-mTOR-p70S6K pathway. Dysregulation of ERs and cell cycle in morusin exposed GBM cells were confirmed by RNA-seq analysis. Finally, we demonstrated the combination of morusin and TMZ remarkably enhanced ER stress and displayed a synergistic effect in GBM cells, and suppressed tumor progression in an orthotopic xenograft model. In conclusion, these findings reveal the toxicity of morusin to GBM cells and its ability to enhance drug sensitivity to TMZ, suggesting the potential application value of morusin in the development of therapeutic strategies for human GBM.

8.
Neuro Oncol ; 24(9): 1482-1493, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35287174

RESUMO

BACKGROUND: Glioblastoma stem cells (GSCs) and their interplay with tumor-associated macrophages (TAMs) are responsible for malignant growth and tumor recurrence of glioblastoma multiforme (GBM), but the underlying mechanisms are largely unknown. METHODS: Cell viability, stemness, migration, and invasion were measured in GSCs after the knockdown of upstream stimulating factor 1 (USF1). Luciferase assay and chromatin immunoprecipitation qPCR were performed to determine the regulation of CD90 by USF1. Immunohistochemistry and immunofluorescent staining were used to examine the expression of USF1 and GSC markers, as well as the crosstalk between GSCs and TAMs. In addition, the interaction between GSCs and TAMs was confirmed using in vivo GBM models. RESULTS: We show that USF1 promotes malignant glioblastoma phenotypes and GSCs-TAMs physical interaction by inducing CD90 expression. USF1 predicts a poor prognosis for glioma patients and is upregulated in patient-derived GSCs and glioblastoma cell lines. USF1 overexpression increases the proliferation, invasion, and neurosphere formation of GSCs and glioblastoma cell lines, while USF1 knockdown exerts an opposite effect. Further mechanistic studies reveal that USF1 promotes GSC stemness by directly regulating CD90 expression. Importantly, CD90 of GSCs functions as an anchor for physical interaction with macrophages. Additionally, the USF1/CD90 signaling axis supports the GSCs and TAMs adhesion and immunosuppressive feature of TAMs, which in turn enhance the stemness of GSCs. Moreover, the overexpression of CD90 restores the stemness property in USF1 knockdown GSCs and its immunosuppressive microenvironment. CONCLUSIONS: Our findings indicate that the USF1/CD90 axis might be a potential therapeutic target for the treatment of glioblastoma.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/genética , Glioblastoma/patologia , Glioma/patologia , Humanos , Células-Tronco Neoplásicas/metabolismo , Antígenos Thy-1/metabolismo , Microambiente Tumoral , Macrófagos Associados a Tumor , Fatores Estimuladores Upstream/metabolismo
9.
Comb Chem High Throughput Screen ; 25(6): 1040-1046, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33797361

RESUMO

OBJECTIVE: The goal of this study was to investigate the status of FEN1 in colorectal cancer (CRC) and determine the potential correlation between FEN1 expression level and clinicopathological parameters in CRC patients. METHODS: Expression of FEN1 in CRC tissue on tissue microarray was detected using immunohistochemistry (IHC). The relationship between FEN1 expression status and clinicopathologic characteristics of CRC was analyzed by the Chi-square test. The survival data of TCGA Colon Cancer (COAD) were obtained from ucsc xena browser (https://xenabrowser.net/). Patients were separated into higher and lower expression groups by median FEN1 expression. The association with prognosis of CRC patients was determined by Kaplan-Meier survival analysis with Log-rank test. RESULTS: FEN1expression level and cellular localization had wide variability among different individuals; we classified the staining results into four types: both positive in nucleus and cytoplasm, both negative in nucleus and cytoplasm, only positive in the nucleus, only positive in the cytoplasm. Moreover, FEN1 expression status only correlated with patient's metastasis status, and the patients in the NLCL group showed more risk of cancer cell metastasis. CONCLUSION: Our results indicate that FEN1 expression level and cellular localization had wide variability in CRC and is not a promising biomarker in CRC.


Assuntos
Neoplasias Colorretais , Biomarcadores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Endonucleases Flap , Humanos , Estimativa de Kaplan-Meier
10.
Front Pharmacol ; 12: 747223, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34938180

RESUMO

Pulmonary fibrosis (PF) is a severe respiratory disease caused by lung microenvironment changes. TGF-ß/Smad3 signaling pathway plays a critical role in the fibrotic process. MicroRNA-29 (miR-29) has proved to alleviate the occurrence of PF by downregulating TGF-ß/Smad3 signaling pathway. The miRNA application encounters obstacles due to its low stability in body and no targeting to lesions. Exosomes can be used for therapeutic delivery of miRNA due to their favorable delivery properties. However, low efficiency of separation and production impedes the therapeutic application of exosomes. In this study, we developed a liquid natural extracellular matrix (ECM) enriched with miR-29-loaded exosomes for PF treatment. The collagen-binding domain (CBD)-fused Lamp2b (CBD-Lamp2b) and miR-29 were overexpressed in human foreskin fibroblast (HFF) host cells for the entrapment of miR-29-loaded exosomes in ECM of the cells. The repeated freeze-thaw method was performed to prepare the liquid ECM enriched with exosomes without destroying the exosomal membrane. In summary, this study developed a novel functional ECM biomaterial for therapy of PF, and also provided a promising gene therapy platform for different diseases by treatment with liquid ECM that is, enriched with exosomes loaded with different functional miRNAs.

11.
Front Genet ; 12: 723725, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759954

RESUMO

Numerous studies have identified various prognostic long non-coding RNAs (LncRNAs) in a specific cancer type, but a comprehensive pan-cancer analysis for prediction of LncRNAs that may serve as prognostic biomarkers is of great significance to be performed. Glioblastoma multiforme (GBM) is the most common and aggressive malignant adult primary brain tumor. There is an urgent need to identify novel therapies for GBM due to its poor prognosis and universal recurrence. Using available LncRNA expression data of 12 cancer types and survival data of 30 cancer types from online databases, we identified 48 differentially expressed LncRNAs in cancers as potential pan-cancer prognostic biomarkers. Two candidate LncRNAs were selected for validation in GBM. By the expression detection in GBM cell lines and survival analysis in GBM patients, we demonstrated the reliability of the list of pan-cancer prognostic LncRNAs obtained above. By constructing LncRNA-mRNA-drug network in GBM, we predicted novel drug-target interactions for GBM correlated LncRNA. This analysis has revealed common prognostic LncRNAs among cancers, which may provide insights into cancer pathogenesis and novel drug target in GBM.

12.
Cell Death Dis ; 12(9): 827, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34480020

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive brain tumor, with a 5-year survival ratio <5%. Invasive growth is a major determinant of the poor prognosis in GBM. In this study, we demonstrate that high expression of PPFIA binding protein 1 (PPFIBP1) correlates with remarkable invasion and poor prognosis of GBM patients. Using scratch and transwell assay, we find that the invasion and migration of GBM cells are promoted by overexpression of PPFIBP1, while inhibited by knockdown of PPFIBP1. Then, we illustrate that overexpression of PPFIBP1 facilitates glioma cell infiltration and reduces survival in xenograft models. Next, RNA-Seq and GO enrichment analysis reveal that PPFIBP1 regulates differentially expressed gene clusters involved in the Wnt and adhesion-related signaling pathways. Furthermore, we demonstrate that PPFIBP1 activates focal adhesion kinase (FAK), Src, c-Jun N-terminal kinase (JNK), and c-Jun, thereby enhancing Matrix metalloproteinase (MMP)-2 expression probably through interacting with SRCIN1 (p140Cap). Finally, inhibition of phosphorylation of Src and FAK significantly reversed the augmentation of invasion and migration caused by PPFIBP1 overexpression in GBM cells. In conclusion, these findings uncover a novel mechanism of glioma invasion and identify PPFIBP1 as a potential therapeutic target of glioma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Glioma/patologia , Sistema de Sinalização das MAP Quinases , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Linhagem Celular Tumoral , Adesões Focais/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/diagnóstico por imagem , Glioma/genética , Células HEK293 , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Fosforilação , Prognóstico , Ligação Proteica , Proteínas Proto-Oncogênicas c-jun/metabolismo , Análise de Sobrevida , Regulação para Cima/genética , Cicatrização
13.
Neoplasma ; 68(1): 126-134, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32880467

RESUMO

Glioma is the most common type of brain cancer. Chemotherapy combination with surgery and radiotherapy is a standard treatment for patients. Although there are many advances in glioma therapy, the prognosis of glioma patients has not significantly been improved over the past decades. Hence, there is still an urgent need to develop a new therapy to treat glioma. Cell viability was assessed by CellTiter Blue assay; flow cytometry (FCM) was used for detecting cell apoptosis; ROS detection was detected by ROS Assay; H2O2 detection was performed by hydrogen peroxide detection kits; real-time PCR and WB were used to determine gene expression. Using the glioma cell line U251 and U87, we investigated a possible combination inhibitory effect includes metformin and cold atmospheric plasma (CAP). The combination treatment showed a synergistic inhibitory effect on cell viability, significantly inducing cell apoptosis. Furthermore, we also found H2O2 produced by CAP has an important role in the synergistic inhibitory effect, eliminating H2O2 with catalase reversed the synergistic inhibitory effect. In addition, the transcript and protein levels of c-FOS were robustly increased after co-treated with metformin and CAP. Taken together, we propose that pre-treatment of glioma cells with metformin sensitize tumor cells to CAP, which may serve as a potential therapeutic strategy for glioma.


Assuntos
Neoplasias Encefálicas , Glioma , Metformina , Gases em Plasma , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glioma/tratamento farmacológico , Glioma/patologia , Glioma/terapia , Humanos , Peróxido de Hidrogênio , Metformina/farmacologia , Gases em Plasma/farmacologia
14.
Front Bioeng Biotechnol ; 9: 816089, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35155414

RESUMO

Cell-laden hydrogel microspheres with uniform size show great potential for tissue repair and drug screening applications. Droplet microfluidic systems have been widely used for the generation of cell-laden hydrogel microspheres. However, existing droplet microfluidic systems are mostly based on complex chips and are not compatible with well culture plates. Moreover, microspheres produced by droplet microfluidics need demulsification and purification from oil, which requires time and effort and may compromise cell viability. Herein, we present a simple one-step approach for producing and purifying hydrogel microspheres with an easily assembled microfluidic device. Droplets were generated and solidified in the device tubing. The obtained hydrogel microspheres were then transferred to a tissue culture plate filled with cell culture media and demulsified through evaporation of the oil at 37°C. The removal of oil caused the gelled microspheres to be released into the cell culture media. The encapsulated cells demonstrated good viability and grew into tumor spheroids in 12-14 days. Single cell-laden hydrogel microspheres were also obtained and grown into spheroid in 14 days. This one-step microsphere generation method shows good potential for applications in automated spheroid and organoid cultures as well as drug screening, and could potentially offer benefits for translation of cell/microgel technologies.

15.
Anticancer Drugs ; 31(10): 1012-1017, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33009034

RESUMO

Tumor cells can activate platelets, which in turn facilitate tumor cell survival and dissemination. Platelets inhibition or blocking platelet-tumor cell interactions has become a strategy to suppress tumor progression. In this study, we investigated the effect of ticagrelor, a new antiplatelet drug, on tumor cell proliferation and metastasis. Our results show that ticagrelor not only inhibits the proliferation, migration, and invasion of B16F10 and Lewis lung carcinoma cells but also induces platelet apoptosis. In addition, we find that apoptosis of the platelet cells is dose dependent. Further, the result of in-vivo experiments proved that ticagrelor treatment decreased the tumor metastasis. The results of this study demonstrate that ticagrelor may be a potential anti-tumor agent for tumor metastasis.


Assuntos
Antineoplásicos/farmacologia , Plaquetas/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Inibidores da Agregação Plaquetária/farmacologia , Ticagrelor/farmacologia , Animais , Apoptose/efeitos dos fármacos , Plaquetas/patologia , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Masculino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
16.
Aging (Albany NY) ; 12(20): 20163-20183, 2020 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-33099538

RESUMO

Osteoarthritis (OA) is one of the most painful and widespread chronic degenerative joint diseases and is characterized by destructed articular cartilage and inflamed joints. Previously, our findings indicated that circular RNA ciRS-7 (ciRS-7)/microRNA 7 (miR-7) axis is abnormally expressed in OA, and regulates proliferation, inflammatory responses, and apoptosis of interleukin-1ß (IL-1ß)-stimulated chondrocytes. However, its underlying role in OA remains unknown. In this study, we first validated cartilage degradation and defection of autophagy in samples of OA patients. IL-1ß initially stimulated autophagy of chondrocytes, and ultimately significantly suppressed autophagy. Upregulated ciRS-7/down-regulated miR-7 aggravated IL-1ß-induced cartilage degradation, and restrained autophagy in vitro. Gene sequencing and bioinformatics analysis performed on a control group, IL-1ß group, and IL-1ß+miR-7-mimics group demonstrated that seven of the most significant mRNA candidates were enriched in the interleukin-17 (IL-17) signaling pathway. Increased IL-17A levels were also observed by qRT-PCR and ELISA. In addition, it was revealed that the ciRS-7/miR-7 axis ameliorated cartilage degradation and defection of autophagy by PI3K/AKT/mTOR activation in IL-1ß-induced chondrocytes. Furthermore, an OA model was established in rats with medial meniscus destabilization. miR-7-siRNA-expressing lentiviruses alleviated surgical resection-induced cartilage destruction of OA mice, whereas miR-7 mimics worsened the effects. Thus, these findings revealed that the mechanism of the ciRS-7/miR-7 axis involved regulating OA progression and provided valuable directions for OA treatment.


Assuntos
Autofagia , Cartilagem Articular/enzimologia , Condrócitos/enzimologia , Interleucina-17/metabolismo , MicroRNAs/metabolismo , Osteoartrite/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia/efeitos dos fármacos , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Estudos de Casos e Controles , Linhagem Celular , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Interleucina-17/genética , Interleucina-1beta/farmacologia , Masculino , MicroRNAs/genética , Osteoartrite/genética , Osteoartrite/patologia , RNA Longo não Codificante/genética , Ratos Sprague-Dawley , Transdução de Sinais , Transcriptoma
17.
Mol Omics ; 15(6): 459-469, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31755891

RESUMO

Accumulating evidence has highlighted the important roles of long non-coding RNAs (lncRNAs) acting as competing endogenous RNAs (ceRNAs) in Alzheimer's disease (AD). In this study, we constructed an AD-derived lncRNA-associated ceRNA network (LncACeNET) based on the ceRNA hypothesis and co-expressed correlation analysis of RNAs (miRNAs, mRNAs and lncRNAs) from AD patients. Based on this network, we preliminarily identified new potential AD biomarkers including hsa-miR-155-5p, CERS6-AS1, and CTB-89H12.4. The functional enrichment analysis demonstrated that these inferred biomarkers were significantly correlated with AD-related biological processes such as neuron projection development and neuron projection morphogenesis. Notably, lncRNA CTB-89H12.4 is significantly associated with "calcium ion-regulated exocytosis of neurotransmitter", "chemical synaptic transmission", "presynaptic membrane assembly", "receptor localization to synapse", and "learning". This indicates the important role of CTB-89H12.4 as a promising target for AD therapy. Subsequently, we used the computational pipeline DTINet and discovered 19 lines of probable therapeutic relationships between FDA-approved drugs and CTB-89H12.4, which offered a new avenue to repurpose existing FDA-approved drugs for AD indication. Our study provides a new landscape for LncACeNET in AD, and will benefit mechanism study and new drug development for AD.


Assuntos
Doença de Alzheimer/genética , Biologia Computacional/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Algoritmos , Descoberta de Drogas/métodos , Humanos , Anotação de Sequência Molecular , Software , Navegador
18.
Am J Transl Res ; 11(4): 2359-2369, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31105842

RESUMO

Uterine fibroid is one of the most common solid tumors occurring in reproductive age women. Lack of accurate methods for In vivo quantitative assessment of uterine fibroid progression severely impedes the basic research and drug screen of this disease. To solve this problem, the correlation between bioluminescence imaging (BLI) and initial cell number used to form xenograft was investigated in this study. The results showed that both subcutaneous (SC) and intraperitoneal (IP) D-luciferin administration led to fast increase of bioluminescence signal (BLS) intensity and caused large variation of peak signal intensity of xenografts through the analysis of BLI kinetic curves. We found that a distinct linear stage appeared in xenograft BLI curve for each mouse subjected to IP-injection of D-luciferin. Moreover, a high positive correlation was found between linear slope and the initial number of human uterine fibroid smooth muscle cells (fSMCs) used for xenograft formation. Our research indicates that the slope of linear stage in BLI curve is more appropriate for in vivo quantitative assessment of human uterine fibroid xenograft.

19.
IUBMB Life ; 71(2): 183-191, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30376610

RESUMO

Extracellular matrix (ECM)-independent survival is an essential prerequisite for tumor metastasis, and a hallmark of epithelial cancer stem cells and epithelial-mesenchymal transition (EMT). Here, we found that loss of TP53I11 enhanced, and overexpression of TP53I11 suppressed the ECM-independent survival, EMT, and migration in MCF10A cells. TP53I11 has long been considered as a transcriptional target of TP53. However, we found that TP53I11 regulated the ECM-independent survival by a TP53-independent way. As a metabolic sensor, AMPK promoted anoikis resistance by inhibiting AKT/m-TOR/p70S6K signaling pathway. It was recently revealed that the reciprocal inhibitory relationship between AKT and AMPK regulated adaptation of cells to ECM-detachment. Our results demonstrated that loss of TP53I11 promoted the activation of AKT/m-TOR pathway, increased PGC-1α expression and thereby enhanced OXPHOS in attach-cultured MCF10A cells, but promoted AMPK activation to inhibit AKT/m-TOR/p70S6K signaling pathway in detach-cultured MCF10A cells. This indicates that TP53I11 functions as a mediator to balance activation of AKT and AMPK to adapt cells to different cellular contexts such as ECM-attachment and -detachment. © 2018 IUBMB Life, 71(1):183-191, 2019.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Transição Epitelial-Mesenquimal/genética , Matriz Extracelular/metabolismo , Proteínas de Neoplasias/genética , Poli(ADP-Ribose) Polimerase-1/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Adesão Celular , Linhagem Celular , Movimento Celular , Sobrevivência Celular , Claudina-1/genética , Claudina-1/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Matriz Extracelular/química , Regulação da Expressão Gênica , Humanos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Proteínas de Neoplasias/deficiência , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Vimentina/genética , Vimentina/metabolismo
20.
Oncogene ; 37(47): 6152-6165, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29995872

RESUMO

Epithelial cells aggregate into spheroids when deprived of matrix, and the proclivity for spheroid formation and survival is a hallmark of normal and tumorigenic mammary stem cells. We show here that Nuclear Respiratory Factor 1 (NRF1) is a spheroid promoter by in silico identification of this transcription factor as highly connected to top shRNA-hits deduced from re-iterative selections for shRNAs enriched in MCF10A spheroids. NRF1-promoted spheroid survival is linked to its stimulation of mitochondrial OXPHOS, cell migration, invasion, and mesenchymal transition. Conversely, NRF1 knockdown in breast cancer MDA-MB-231 cells reduced spheroids, migration, invasion, and mesenchymal marker expression. NRF1 knockdown also reduced tumor burden in mammary fat pads and lungs of orthotopic- or tail vein-transplanted mice. With the Luminal A subtype of breast cancer, higher NRF1 expression is associated with lower survival. These results show that NRF1, an activator of mitochondrial metabolism, supports mammary spheroid survival and tumor development.


Assuntos
Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Glândulas Mamárias Humanas/metabolismo , Fator 1 Nuclear Respiratório/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...