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.
J Immunother ; 47(8): 279-302, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38980088

RESUMO

We evaluated the BYSL content and underlying mechanism in melanoma (SKCM) overall survival (OS). In this study, we used a comprehensive approach combining bioinformatics tools, including miRNA estimation, quantitative real-time polymerase chain reaction (qRT-PCR) of miRNAs, E3 ligase estimation, STRING analysis, TIMER analysis, examination of associated upstream modulators, protein-protein interaction (PPI) analysis, as well as retrospective and survival analyses, alongside clinical sample validation. These methods were used to investigate the content of BYSL, its methylation status, its relation to patient outcome, and its immunologic significance in tumors. Our findings revealed that BYSL expression is negatively regulated by BYSL methylation. Analysis of 468 cases of SKCM RNA sequencing samples demonstrated that enhanced BYSL expression was associated with higher tumor grade. We identified several miRNAs, namely hsa-miR-146b-3p, hsa-miR-342-3p, hsa-miR-511-5p, hsa-miR-3690, and hsa-miR-193a-5p, which showed a strong association with BYSL levels. Furthermore, we predicted the E3 ubiquitin ligase of BYSL and identified CBL, FBXW7, FZR1, KLHL3, and MARCH1 as potential modulators of BYSL. Through our investigation, we discovered that PNO1, RIOK2, TSR1, WDR3, and NOB1 proteins were strongly associated with BYSL expression. In addition, we found a close association between BYSL levels and certain immune cells, particularly dendritic cells (DCs). Notably, we observed a significant negative correlation between miR-146b-3p and BYSL mRNA expression in SKCM sera samples. Collectively, based on the previously shown evidences, BYSL can serve as a robust bioindicator of SKCM patient prognosis, and it potentially contributes to immune cell invasion in SKCM.


Assuntos
Biomarcadores Tumorais , Regulação Neoplásica da Expressão Gênica , Melanoma , MicroRNAs , Humanos , Melanoma/mortalidade , Melanoma/genética , Melanoma/diagnóstico , Melanoma/metabolismo , Prognóstico , MicroRNAs/genética , Feminino , Masculino , Biologia Computacional/métodos , Metilação de DNA , RNA Longo não Codificante/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Pessoa de Meia-Idade , Mapas de Interação de Proteínas , Perfilação da Expressão Gênica , Análise de Sobrevida
2.
Front Med (Lausanne) ; 11: 1330491, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38566927

RESUMO

Solar keratosis, also known as actinic keratosis (AK), is becoming increasingly prevalent. It is a benign tumor that develops in the epidermis. Individuals with AK typically exhibit irregular, red, scaly bumps or patches as a result of prolonged exposure to UV rays. These growths primarily appear on sun-exposed areas of the skin such as the face, scalp, and hands. Presently, dermatologists are actively studying AK due to its rising incidence rate in the United States. However, the underlying causes of AK remain poorly understood. Previous research has indicated that the onset of AK involves various mechanisms including UV ray-induced inflammation, oxidative stress, complex mutagenesis, resulting immunosuppression, inhibited apoptosis, dysregulated cell cycle, altered cell proliferation, tissue remodeling, and human papillomavirus (HPV) infection. AK can develop in three ways: spontaneous regression, persistence, or progression into invasive cutaneous squamous cell carcinoma (cSCC). Multiple risk factors and diverse signaling pathways collectively contribute to its complex pathogenesis. To mitigate the risk of cancerous changes associated with long-term UV radiation exposure, prompt identification, management, and prevention of AK are crucial. The objective of this review is to elucidate the primary mechanisms underlying AK malignancy and identify potential treatment targets for dermatologists in clinical settings.

3.
Int J Biol Macromol ; 275(Pt 1): 129705, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38272418

RESUMO

Skin injuries and defects, as a common clinical issue, still cannot be perfectly repaired at present, particularly large-scale and infected skin defects. Therefore, in this work, a drug-loaded bilayer skin scaffold was developed for repairing full-thickness skin defects. Briefly, amoxicillin (AMX) was loaded on polycaprolactone (PCL) nanofiber via electrospinning to form the antibacterial nanofiber membrane (PCL-AMX) as the outer layer of scaffold to mimic epidermis. To maintain wound wettability and promote wound healing, external human epidermal growth factor (rhEGF) was loaded in sodium alginate-gelatin to form the hydrogel structure (SG-rhEGF) via 3D printing as inner layer of scaffold to mimic dermis. AMX and rhEGF were successfully loaded into the scaffold. The scaffold exhibited excellent physicochemical properties, with elongation at break and tensile modulus were 102.09 ± 6.74% and 206.83 ± 32.10 kPa, respectively; the outer layer was hydrophobic (WCA was 112.09 ± 4.67°), while the inner layer was hydrophilic (WCA was 48.87 ± 5.52°). Meanwhile, the scaffold showed excellent drug release and antibacterial characteristics. In vitro and in vivo studies indicated that the fabricated scaffold could enhance cell adhesion and proliferation, and promote skin wound healing, with favorable biocompatibility and great potential for skin regeneration and clinical application.


Assuntos
Alginatos , Antibacterianos , Gelatina , Hidrogéis , Nanofibras , Poliésteres , Impressão Tridimensional , Pele , Alicerces Teciduais , Cicatrização , Gelatina/química , Cicatrização/efeitos dos fármacos , Nanofibras/química , Antibacterianos/farmacologia , Antibacterianos/química , Poliésteres/química , Alginatos/química , Alginatos/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Alicerces Teciduais/química , Pele/efeitos dos fármacos , Animais , Amoxicilina/farmacologia , Amoxicilina/química , Humanos , Liberação Controlada de Fármacos
4.
Biomed Res Int ; 2022: 5532118, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35378790

RESUMO

Aims: Cryptococcosis is an invasive fungal disease that is associated with an increasing prevalence along with a very high fatality and is primarily caused by Cryptococcus. However, its mechanism to cause pathogenicity is not yet completely understood. In this study, we aim to screen the lncRNA markers in human monocytic (THP-1) cells infected by Cryptococcus neoformans (C. neoformans) through high-throughput sequencing technology and to explore its effects on biological functions. Methods: We initially conducted an lncRNA microarray analysis of the THP-1 cells infected by C. neoformans and normal THP-1 cells. Based upon these data, RT-qPCR was used to verify the expressions of the selected lncRNAs and mRNAs. We then performed functional and pathway enrichment analyses. Lastly, target prediction was performed by using the lncRNA target tool which was based on the differentially expressed lncRNAs. Results: We determined 81 upregulated and 96 downregulated lncRNAs using microarray. In addition, the profiling data showed 42 upregulated and 57 downregulated genes and discovered that neuroactive ligand-receptor interaction, tyrosine metabolism, and phenylalanine metabolism are extremely impaired in the regulation of C. neoformans infection. GO enrichment analysis of the 99 differentially expressed mRNAs exhibited that these modules showed different signaling pathways and biological mechanisms like protein binding and metal ion binding. Moreover, lncRNAs and mRNAs were analyzed for their coexpression relations. A qRT-PCR analysis confirmed that the expression of the top 10 differently expressed mRNA and lincRNA. The expressions of the lncRNAs after C. neoformans infection in THP-1 cells were detected by RNA-sequence, suggesting that microarray analysis could reveal lncRNAs having functional significance that might be linked with the progression of patients. Conclusion: The current study analyzed the differential lncRNAs and mRNAs in C. neoformans infection and predicted the corresponding pathways and their correlations that can offer new potential insights into the mechanistic basis of this condition.


Assuntos
Criptococose , RNA Longo não Codificante , Criptococose/genética , Cryptococcus neoformans , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células THP-1
5.
Int J Mol Med ; 48(5)2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34498713

RESUMO

Investigating the factors that influence the inflammatory response of microglial cells is crucial for understanding the pathogenesis of cryptococcal meningitis (CM). MicroRNAs (miRNAs/miRs) play an important role in inducing host defenses and activating the immune response during microbial infection; however, the regulatory mechanisms of miRNAs in cryptococcal meningitis remain poorly defined. In a previous study, the authors assessed the miRNA profiles of THP­1 (human acute monocytic leukemia cells) cells following Cryptococcus neoformans (C. neoformans) infection. In the present study, it was found that miR­4792 expression was downregulated in BV2 cells infected with C. neoformans, whilst that of its target gene, epidermal growth factor receptor (EGFR), was upregulated. Infected cells in which miR­4792 was overexpressed exhibited a decreased EGFR transcript expression, reduced mitogen­activated protein kinase (MAPK) signaling and a decreased secretion of inflammatory cytokines. In addition, following antifungal treatment in patients with cryptococcal meningitis, the levels of miR­4792 in the cerebrospinal fluid significantly increased, whilst the expression of EGFR significantly decreased. In addition, receiver operator characteristic analysis revealed miR­4792 (AUCROC=0.75) and EGFR (AUCROC=0.79) as potential diagnostic markers in patients with cryptococcal meningitis.


Assuntos
Criptococose/genética , Criptococose/microbiologia , Cryptococcus neoformans/fisiologia , Inflamação/genética , MicroRNAs/metabolismo , Microglia/metabolismo , Microglia/microbiologia , Adolescente , Adulto , Animais , Sequência de Bases , Linhagem Celular , Citocinas/biossíntese , Receptores ErbB/metabolismo , Feminino , Humanos , Inflamação/patologia , Masculino , Meningite Criptocócica/imunologia , Meningite Criptocócica/microbiologia , Camundongos , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células THP-1 , Adulto Jovem
6.
Microb Pathog ; 141: 103959, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31958475

RESUMO

Cryptococcosis is a disease predominantly caused by Cryptococcus neoformans in China and C. neoformans is the main form that causes cryptococcal meningitis. In this study, we examined the influence of MiR-30c-5p during Cryptococcus neoformans infection. microRNAs were extracted from Cerebrospinal fluid and sera of patients. To identify pathogenic microRNAs, RNASeq were performed. The results were confirmed with quantitative real-time PCR (qRT-PCR), transient transfection of siRNAs or microRNA mimics into cultured BV2 cell, flow cytometry, immunoblotting, luciferase assay and immunohistochemistry. In this study we found that miR-30c expression was downregulated and that inflammation, apoptosis, and autophagy were activated. The overexpression of miR-30c-5p significantly inhibited inflammation and autophagic activity and decreased apoptosis, and treatment with sieIF2α resulted in a significant decrease in inflammation, apoptosis. In addition, clinical samples of cerebrospinal fluid and serum of patients with cryptococcal meningitis who have undergone standard antifungal treatment showed that the expression of miR-30c-5p was increased while that of eIF2α was decreased, which was in accordance with the in vitro experiments. These studies demonstrated that miRNA-30c-5p can inhibit inflammatory, apoptotic, and autophagic activity through the eIF2α/ATF4 pathway, and it is thus a potential target for the diagnosis, treatment, and detection of cryptococcal meningitis.


Assuntos
Criptococose/genética , Criptococose/microbiologia , Fator de Iniciação 2 em Eucariotos/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Microglia/metabolismo , Interferência de RNA , Adolescente , Adulto , Animais , Apoptose/genética , Autofagia/genética , Biomarcadores , Linhagem Celular , Criptococose/imunologia , Criptococose/metabolismo , Cryptococcus neoformans , Citocinas/metabolismo , Feminino , Genes Reporter , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Masculino , Camundongos , Microglia/patologia , Microglia/ultraestrutura , Pessoa de Meia-Idade , Transdução de Sinais , Adulto Jovem
7.
Eur J Dermatol ; 29(4): 359-365, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31625918

RESUMO

BACKGROUND: Previous reports have shown that epithelial-to-mesenchymal transition (EMT) indicates the importance of transforming growth factor-ß (TGF-ß) signalling in the pathogenesis of systemic sclerosis (SSc). However, the underlying molecular mechanisms of EMT are not fully understood. OBJECTIVES: Brachyury, an evolutionarily conserved transcription factor, was recently identified as an important factor that promotes EMT in human carcinoma cell lines. However, there is no evidence indicating that brachyury is involved in EMT in SSc. MATERIALS AND METHODS: The expression of brachyury and collagen was investigated in cultures of dermal fibroblasts and skin sections derived from SSc patients and healthy controls. Brachyury and collagen expression were determined by immunohistochemistry and immunoblotting, respectively, and mRNA for both was analysed using real-time PCR. RESULTS: Brachyury was overexpressed in SSc dermal fibroblasts both in vivo and in vitro, and this overexpression was inhibited by TGF-ß1 inhibitor. Brachyury siRNA reduced mRNA and protein expression levels of type I collagen in normal and SSc dermal fibroblasts, but did not decrease the levels of major disease-related cytokines. Furthermore, brachyury levels were significantly increased in skin samples of SSc patients relative to healthy controls. CONCLUSIONS: The up-regulation of brachyury in response to activated endogenous TGF-ß signalling may play a role in constitutive up-regulation of collagen in SSc fibroblasts. Further studies assessing the regulatory mechanism of tissue fibrosis induced by brachyury in SSc skin may lead to a better understanding of the pathogenesis, new diagnostic methods, and new therapeutic approaches using siRNAs.


Assuntos
Colágeno Tipo I/genética , Transição Epitelial-Mesenquimal/genética , Proteínas Fetais/genética , Escleroderma Sistêmico/genética , Proteínas com Domínio T/genética , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Biópsia por Agulha , Estudos de Casos e Controles , Células Cultivadas , Feminino , Fibroblastos/citologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência , Escleroderma Sistêmico/patologia , Transdução de Sinais/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA