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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Microb Pathog ; 140: 103943, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31917273

RESUMO

BACKGROUND: The impact of NBUVB on the cutaneous microbiota of vitiligo patients remains to be fully elucidated. METHODS: To characterize the cutaneous microbiota in vitiligo patients, cutaneous samples from 60 patients with vitiligo and after NBUVB irradiation were profiled using the Illumina MiSeq platform. Alpha diversity estimations revealed higher microbiota diversity in samples from patients with lesional skin. Beta diversity (Principal Component Analysis (PCA)) analysis showed that the bacterial community structure segregated differently between different groups. RESULTS: There was a statistically significant increase in the Sobs, ACE, and Chao indices in the NB group compared with NF group, as determined by t-test. The alpha diversity have no significant difference between NF and DB group. At the phylum level, Firmicutes, Proteobacteria and Actinobacteria were the most predominant phyla. Propionibacterium and Pseudomonas were the most predominant genera in each group. In addition, Staphylococcus, Bacillus and Prevotella were enriched in DF group compared to DB group. Propionibacterium was enriched in DB group compared to DF group. CONCLUSIONS: Our studies indicate differences in microbial community dynamics of the lesional and non-lesional sites of vitiligo subjects, with greater diversity and higher association between microbial communities of the unaffected site. And NBUVB irradiation might eliminate these differences.


Assuntos
Microbiota/genética , Pele/microbiologia , Terapia Ultravioleta , Vitiligo/microbiologia , Actinobacteria/classificação , Actinobacteria/genética , Actinobacteria/isolamento & purificação , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Feminino , Firmicutes/classificação , Firmicutes/genética , Firmicutes/isolamento & purificação , Humanos , Masculino , Metagenômica , Proteobactérias/classificação , Proteobactérias/genética , Proteobactérias/isolamento & purificação , RNA Ribossômico 16S , Pele/patologia
2.
Dermatol Ther ; 32(5): e13025, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31306558

RESUMO

Our study aimed to identify the key genes and upstream regulators in vitiligo. To screen the pathogenic genes of vitiligo, an integrated analysis was performed by using the microarray datasets in vitiligo derived from the Gene Expression Omnibus (GEO) database. The functional annotation and potential pathways of differentially expressed genes (DEGs) were further explored by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. We constructed a vitiligo-specific transcriptional regulatory network to identify crucial transcriptional factors that target the DEGs in vitiligo. From two GEO datasets, we identified 1863 DEGs (744 downregulated DEGs and 1,119 upregulated DEGs [false discovery rate < 0.05, |Combined.ES| > 1]) between lesional tissues and nonlesional tissues. GO and KEGG analyses revealed that ubiquitin-mediated proteolysis and the endoplasmic reticulum were significantly enriched pathways for DEGs. The expressions of premelanosome (PMEL), melan-A (MLANA), dopachrome tautomerase (DCT), SRY-boxtranscription factor 10 (SOX10), tyrosinase-related protein 1 (TYRP1), and melanocortin 1 receptor (MC1R) were shown to be involved in the pathogenesis of vitiligo. We concluded that PMEL, MLANA), DCT, SOX10, TYRP1, and MC1R may play a role in vitiligo, among which TYRP1 and MC1R are regulated by forkhead box J2 (FOXJ2). Our finding may contribute to the development of new potential biomarkers, reveal the underlying pathogenesis of vitiligo, and identify novel therapeutic targets for vitiligo.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , RNA/genética , Fatores de Transcrição/genética , Vitiligo/genética , Ontologia Genética , Marcadores Genéticos/genética , Humanos , Transdução de Sinais , Fatores de Transcrição/metabolismo , Vitiligo/metabolismo
3.
Med Sci Monit ; 22: 5141-5153, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-28025572

RESUMO

BACKGROUND We designed and carried out this study to examine the role of miR-155 and the Rheb/mTOR pathway in ischemic stroke. We also investigated how these two elements interact with each other and contribute to injuries resulting from ischemic stroke. MATERIAL AND METHODS We used both a middle cerebral artery occlusion rat model in vivo and an oxygen-glucose deprivation cell model in vitro to simulate the onset of ischemic stroke. miR-155 mimics, miR-155 inhibitors, and Rheb siRNA were transfected to alter the expression of miR-155 and Rheb. Infarct sizes were measured using magnetic resonance imaging (MRI) and triphenyltetrazolium chloride (TTC) staining; cell apoptosis rates were calculated using Annexin V-FITC/PI staining and flow cytometry. Levels of miR-155, Rheb, mTOR, and S6K were examined by RT-PCR, immunofluorescence, and western blot. We performed a luciferase activity assay so that the association between miR-155 and Rheb could be fully assessed. RESULTS We demonstrated that miR-155 bound the 3'-UTR of Rheb and suppressed Rheb expression. As suggested by animal models, significant cerebral infarct volumes and cell apoptosis were induced by increased expression of miR-155 and decreased expression of Rheb, mTOR, and p-S6K (P<0.05). miR-155 inhibitors exhibited protective effects on ischemic stroke, including down-regulation of infarction size in cerebral tissues in vivo and reduced apoptosis of BV2 cells in vitro with increased expression of Rheb, mTOR and p-S6K (P<0.05). These protective effects could be substantially antagonized by the transfection of Rheb siRNA (P<0.05). CONCLUSIONS Inhibition of miR-155 may play protective roles in ischemic stroke by phosphorylating S6K through the Rheb/mTOR pathway.


Assuntos
Apoptose/genética , Isquemia Encefálica/genética , Encéfalo/metabolismo , MicroRNAs/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/genética , Serina-Treonina Quinases TOR/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Sítios de Ligação , Encéfalo/patologia , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Linhagem Celular , Modelos Animais de Doenças , Citometria de Fluxo , Imuno-Histoquímica , Luciferases/metabolismo , Imageamento por Ressonância Magnética , Camundongos , Fosforilação , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Coloração e Rotulagem , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia , Transfecção
4.
J Dermatol Sci ; 100(3): 201-208, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33127205

RESUMO

BACKGROUND: JAK2/STAT3 pathway is involved in the development and progression of melanoma once DNA damage is caused by environment and genetic factors. OBJECTIVE: Here, we aimed to identify novel inhibitor of JAK2/STAT3 pathway and reveal the underlying mechanisms. METHODS: Eighty MedChemExpress compounds were screened by using STAT3-Luc reporter in A375 cells. Podocarpusflavone A (PCFA) was identified as an inhibitor of STAT3, which was further verified in four melanoma cell lines. The anti-melanoma effects and mechanism of PCFA were examined and explored in melanoma cells and mouse xenograft models by using Western blot and cell-counting kit-8 assay. RESULTS: PCFA exhibited potent inhibitory effects on melanoma both in vitro and in vivo. PCFA inhibited the activation of STAT3 through suppressing the phosphorylation of JAK2, and then restrained cell cycle and induced apoptosis of melanoma cells. CONCLUSION: PCFA inhibits melanoma growth via the inhibition of JAK2/STAT3 pathway, which provides a promising therapeutic strategies of melanoma treatment.


Assuntos
Flavonoides/farmacologia , Melanoma/tratamento farmacológico , Fator de Transcrição STAT3/antagonistas & inibidores , Neoplasias Cutâneas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Flavonoides/uso terapêutico , Humanos , Janus Quinase 2/metabolismo , Masculino , Melanoma/patologia , Camundongos , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncol Lett ; 14(1): 635-638, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28693215

RESUMO

We investigated the expression of FOXC2, PinX1, Ki-67 and Cyclin D1 in cutaneous cell carcinoma. We collected 30 cutaneous squamous cell carcinoma (SCC), 30 cutaneous basal cell carcinoma (BCC) and 30 normal skin tissues. The protein expression and gene expression of FOXC2, endogenous telomerase-inhibiting gene PinX1, Ki-67 and Cyclin D1 was measured by immunohistochemistry (IHC) and semi-quantitative reverse transcription polymerase chain reaction (RT-PCR), respectively. In SCC and BCC tissues, the positive rate of protein expression and mRNA level of PinX1 were both significantly lower than those in normal tissues. However, the positive rate of protein expression and mRNA level of FOXC2, Ki-67 and Cyclin D1 were significantly higher than those in normal tissues (p<0.05). There was no significant difference between SCC and BCC (p>0.05). In conclusion, FOXC2 may participate in the carcinogenesis process of SCC and BCC. It may also correlate with the expression PinX, Ki-67 and Cyclin D1. However, FOXC2 alone cannot be used as a diagnostic indicator of SCC.

6.
Biosci Rep ; 37(3)2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28487474

RESUMO

Long noncoding RNAs (lncRNAs) are frequently dysregulated and have important roles in many diseases, particularly cancers. lncRNA-HEIH was first identified in hepatocellular carcinoma (HCC). The expression, clinical significance and roles of lncRNA-HEIH in melanoma are still unknown. In the present study, we found that lncRNA-HEIH is highly expressed in melanoma tissues and cell lines, associated with advanced clinical stages, and predicts poor outcomes in melanoma patients. Functional assays showed that ectopic expression of lncRNA-HEIH promotes melanoma cell proliferation, migration and invasion. Knockdown of lncRNA-HEIH inhibits melanoma cell proliferation, migration and invasion. Mechanistically, we revealed that lncRNA-HEIH directly binds to miR-200b/a/429 promoter and represses miR-200b/a/429 transcription. The expression of miR-200b is inversely associated with lncRNA-HEIH in melanoma tissues. Furthermore, overexpression of miR-200b/a/429 abrogates melanoma cell proliferation, migration and invasion enhanced by lncRNA-HEIH. In conclusion, we identified lncRNA-HEIH as a key oncogene in melanoma via transcriptional inhibition of miR-200b/a/429 Our data suggested that lncRNA-HEIH may serve as a promising prognostic biomarker and therapeutic target for melanoma.


Assuntos
Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanoma/patologia , MicroRNAs/genética , RNA Longo não Codificante/metabolismo , Neoplasias Cutâneas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo/genética , Humanos , Estimativa de Kaplan-Meier , Melanócitos , Melanoma/metabolismo , Invasividade Neoplásica , Regiões Promotoras Genéticas , Neoplasias Cutâneas/metabolismo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA