Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ann Nucl Med ; 37(10): 541-550, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37422857

RESUMO

OBJECTIVE: The aim of this meta-analysis was to compare the diagnostic value of noninvasive imaging methods computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) in the detection of ovarian cancer (OC). METHODS: PubMed, Embase, and Ovid were comprehensively searched from the date of inception to 31st, March, 2022. Pooled sensitivity, specificity, positive likelihood ratio (+ LR), negative likelihood ratio (- LR), diagnostic odds ratio (DOR), and area under the curve (AUC) of summary receiver operating characteristic (SROC) with their respective 95% confidence intervals (CIs) were calculated. RESULTS: Sixty-one articles including 4284 patients met the inclusion criteria of this study. Pooled estimates of sensitivity, specificity, and AUC of SROC with respective 95% CIs of CT on patient level were 0.83 (0.73, 0.90), 0.69 (0.54, 0.81), and 0.84 (0.80, 0.87). The overall sensitivity, specificity, SROC value with respective 95% CIs of MRI were 0.95 (0.91, 0.97),0.81 (0.76, 0.85), and 0.90 (0.87, 0.92) on patient level. Pooled estimates of sensitivity, specificity, SROC value of PET/CT on patient level were 0.92 (0.88, 0.94), 0.88 (0.83, 0.92), and 0.96 (0.94, 0.97). CONCLUSION: Noninvasive imaging modalities including CT, MRI, PET (PET/CT, PET/MRI) yielded favorable diagnostic performance in the detection of OC. Hybrid implement of different tools (PET/MRI) is more accurate for identifying metastatic OC.


Assuntos
Neoplasias Ovarianas , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Humanos , Feminino , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada por Raios X/métodos , Imageamento por Ressonância Magnética/métodos , Neoplasias Ovarianas/diagnóstico por imagem , Sensibilidade e Especificidade , Fluordesoxiglucose F18
2.
Cells ; 11(15)2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35954307

RESUMO

The static magnetic fields (SMFs) impact on biological systems, induce a variety of biological responses, and have been applied to the clinical treatment of diseases. However, the underlying mechanisms remain largely unclear. In this report, by using human mesenchymal stem cells (MSCs) as a model, we investigated the biological effect of SMFs at a molecular and cellular level. We showed that SMF exposure promotes MSC proliferation and activates the expression of transcriptional factors such as FOS (Fos Proto-Oncogene, AP-1 Transcription Factor Subunit) and EGR1 (Early Growth Response 1). In addition, the expression of signal-transduction proteins p-ERK1/2 and p-JNK oscillate periodically with SMF exposure time. Furthermore, we found that the inhibition of the T-type calcium ion channels negates the biological effects of SMFs on MSCs. Together, we revealed that the SMFs regulate T-type calcium ion channels and mediate MSC proliferation via the MAPK signaling pathways.


Assuntos
Canais de Cálcio Tipo T , Células-Tronco Mesenquimais , Canais de Cálcio Tipo T/metabolismo , Proliferação de Células , Humanos , Sistema de Sinalização das MAP Quinases , Campos Magnéticos , Células-Tronco Mesenquimais/metabolismo
3.
Sci China Life Sci ; 65(8): 1481-1497, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35696016

RESUMO

The CREB1 gene encodes an exceptionally pleiotropic transcription factor that frequently dysregulated in human cancers. CREB1 can regulate tumor cell status of proliferation and/or migration; however, the molecular basis for this switch involvement in cell plasticity has not fully been understood yet. Here, we first show that knocking out CREB1 triggers a remarkable effect of epithelial-mesenchymal transition (EMT) and leads to the occurrence of inhibited proliferation and enhanced motility in HCT116 colorectal cancer cells. By monitoring 45 cellular signaling pathway activities, we find that multiple growth-related pathways decline significantly while inflammatory pathways including NF-κB are largely upregulated in comparing between the CREB1 wild-type and knocked out cells. Mechanistically, cells with CREB1 knocked out show downregulation of MYC as a result of impaired CREB1-dependent transcription of the oncogenic lncRNA CCAT1. Interestingly, the unbalanced competition between the coactivator CBP/p300 for CREB1 and p65 leads to the activation of the NF-κB pathway in cells with CREB1 disrupted, which induces an obvious EMT phenotype of the cancer cells. Taken together, these studies identify previously unknown mechanisms of CREB1 in CRC cell plasticity via regulating lncRNA CCAT1 and NF-κB pathways, providing a critical insight into a combined strategy for CREB1-targeted tumor therapies.


Assuntos
Plasticidade Celular , Neoplasias Colorretais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , NF-kappa B , RNA Longo não Codificante , Linhagem Celular Tumoral , Movimento Celular/genética , Plasticidade Celular/genética , Plasticidade Celular/fisiologia , Proliferação de Células/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/fisiopatologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
4.
Phys Rev Lett ; 128(6): 067701, 2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35213197

RESUMO

Manipulating quantum thermal transport relies on uncovering the principle working cycles of quantum devices. Here we introduce the cycle flux ranking of network analysis to nonequilibrium thermal devices characterized as a quantum-transition network. To excavate the principal mechanism out of complex transport behaviors, we decompose the network into cycle trajectories, collect the cycle fluxes by algebraic graph theory, and select top-ranked cycle fluxes, i.e., the cycle trajectories with highest probabilities. We exemplify the cycle flux ranking in typical quantum device models, e.g., a thermal-drag spin-Seebeck pump and a quantum thermal transistor. Top-ranked cycle trajectories indeed elucidate the principal working mechanisms. Therefore, cycle flux ranking provides an alternative perspective that naturally describes the working cycle corresponding to the main functionality of quantum thermal devices, which would further guide the device optimization with desired performance.

5.
Front Oncol ; 12: 954345, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36644638

RESUMO

Background: CD22 single and CD19/CD22 bispecific targeted chimeric antigen receptor T (CAR-T) cell therapy are promising immunotherapy modalities for the treatment of hematologic malignancies. The aim of this study was to assess the efficacy and safety of CD22 and CD19/CD22 targeted CAR-T cell therapy by summarizing the existing evidence. Methods: Electronic databases including PubMed, Embase, and Scopus were comprehensively searched from inception up to November 30, 2022. Pooled response rates and minimal residual disease (MRD) negative response rates, cytokine release syndrome (CRS) rates and neurotoxicity rates were calculated. Subgroup analysis was performed based on the type of immunotherapy. Results: Ten clinical studies including 194 patients with hematologic malignancies were included after a systematical screening of literature. The pooled complete response (CR) rates of CD22 and CD19/CD22 CAR-T cell therapy for relapsed or refractory B-cell lymphoblastic leukemia (B-ALL) were 0.75 (95% CI: 0.60 - 0.88) and 0.87 (95% CI: 0.76 - 0.96). The overall MRD negative response rates of CD22 and CD19/CD22 CAR-T were 0.54 (95% CI: 0.42 - 0.66) and 0.91 (95% CI: 0.47 - 0.88). Pooled CRS rates of CD22 targeted and CD19/CD22 targeted immunotherapy were 0.92 (95% CI: 0.82 - 0.98) and 0.94 (95% CI: 0.82 - 1.00), respectively. Conclusion: Both CD22 and CD19/CD22 CAR-T immunotherapy demonstrated favorable efficacy and acceptable adverse events in the treatment of hematologic malignancies. Well-designed and large sample-sized clinical trials are warranted.

6.
Mol Oncol ; 12(11): 1949-1964, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30171794

RESUMO

miR-372/373, a cluster of stem cell-specific microRNAs transactivated by the Wnt pathway, has been reported to be dysregulated in various cancers, particularly colorectal cancer (CRC); however, the unique role of these microRNAs in cancer remains to be discovered. In the present study, we characterized the upregulation in expression of miR-372/373 in CRC tissues from The Cancer Genome Atlas data, and then showed that overexpression of miR-372/373 enhanced the stemness of CRC cells by enriching the CD26/CD24-positive cell population and promoting self-renewal, chemotherapy resistance and the invasive potential of CRC cells. To clarify the mechanism underlying microRNA-induced stemness, we profiled 45 cell signaling pathways in CRC cells overexpressing miR-372/373 and found that stemness-related pathways, such as Nanog and Hedgehog, were upregulated. Instead, differentiation-related pathways, such as NFκB, MAPK/Erk and VDR, were markedly repressed by miR-372/373. Numerous new targets of miR-372/373 were identified, including SPOP, VDR and SETD7, all of which are factors important for cell differentiation. Furthermore, in contrast to the increase in miR-372/373 expression in CRC tissues, the expression levels of SPOP and VDR mRNA were significantly downregulated in these tissues, indicative of the poor differentiation status of CRC. Taken together, our findings suggest that miR-372/373 enhance CRC cell stemness by repressing the expression of differentiation genes. These results provide new insights for understanding the function and mechanisms of stem cell-specific microRNAs in the development of metastasis and drug resistance in CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , RNA Neoplásico/metabolismo , Animais , Células CACO-2 , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/patologia , RNA Neoplásico/genética
7.
Oncotarget ; 7(27): 42513-42526, 2016 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-27285761

RESUMO

A hallmark of aberrant activation of the Wnt/ß-catenin signaling pathway has been observed in most colorectal cancers (CRC), but little is known about the role of non-coding RNAs regulated by this pathway. Here, we found that miR-150 was the most significantly upregulated microRNA responsive to elevated of Wnt/ß-catenin signaling activity in both HCT116 and HEK293T cells. Mechanistically, the ß-catenin/LEF1 complex binds to the conserved TCF/LEF1-binding element in the miR-150 promoter and thereby transactivates its expression. Enforced expression of miR-150 in HCT116 cell line transformed cells into a spindle shape with higher migration and invasion activity. miR-150 markedly suppressed the CREB signaling pathway by targeting its core transcription factors CREB1 and EP300. Knockdown of CREB1 or EP300 and knockout of CREB1 by CRISPR/Cas9 phenocopied the epithelial-mesenchymal transition (EMT) observed in HCT116 cells in response to miR-150 overexpression. In summary, our data indicate that miR-150 is a novel Wnt effector that may significantly enhance EMT of CRC cells by targeting the CREB signaling pathway.


Assuntos
Transição Epitelial-Mesenquimal , MicroRNAs/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Movimento Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Progressão da Doença , Proteína p300 Associada a E1A/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HEK293 , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ativação Transcricional , Transfecção
8.
Mol Cancer Ther ; 11(5): 1155-65, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22389469

RESUMO

The transcription factor c-Myc is important in cell fate decisions and is frequently overexpressed in cancer cells, making it an attractive therapeutic target. Natural compounds are among the current strategies aimed at targeting c-Myc, but their modes of action still need to be characterized. To explore the mechanisms underlying the anticancer activity of a natural diterpenoid, oridonin, we conducted miRNA expression profiling and statistical analyses that strongly suggested that c-Myc was a potential molecular target of oridonin. Furthermore, experimental data showed that oridonin significantly reduced c-Myc protein levels in vitro and in vivo and that this reduction was mediated by the ubiquitin-proteasome system. Fbw7, a component of the ubiquitin-proteasome system and an E3 ubiquitin ligase of c-Myc, was upregulated rapidly in K562 cells and other leukemia and lymphoma cells, resulting in the rapid turnover of c-Myc. In cell lines harboring mutations in the WD domain of Fbw7, the degradation of c-Myc induced by oridonin was attenuated during short-term treatment. GSK-3, an Fbw7 priming kinase, was also activated by oridonin, along with an increase in T58-phosphorylated c-Myc. Furthermore, the knockdown of Fbw7 or the forced expression of stable c-Myc resulted in reduced sensitization to oridonin-induced apoptosis. Our observations help to clarify the anticancer mechanisms of oridonin and shed light on the application of this natural compound as an Fbw7-c-Myc pathway targeting agent in cancer treatment.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Diterpenos do Tipo Caurano/farmacologia , Proteínas F-Box/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Proteína 7 com Repetições F-Box-WD , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Células HL-60 , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/genética , Fosforilação/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais/efeitos dos fármacos , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...