Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 51(1): 814, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39008163

RESUMO

Periodontitis is a severe gum infection that begins as gingivitis and can lead to gum recession, bone loss, and tooth loss if left untreated. It is primarily caused by bacterial infection, which triggers inflammation and the formation of periodontal pockets. Notably, periodontitis is associated with systemic health issues and has been linked to heart disease, diabetes, respiratory diseases, adverse pregnancy outcomes, and cancers. Accordingly, the presence of chronic inflammation and immune system dysregulation in individuals with periodontitis significantly contributes to the initiation and progression of various cancers, particularly oral cancers. These processes promote genetic mutations, impair DNA repair mechanisms, and create a tumor-supportive environment. Moreover, the bacteria associated with periodontitis produce harmful byproducts and toxins that directly damage the DNA within oral cells, exacerbating cancer development. In addition, chronic inflammation not only stimulates cell proliferation but also inhibits apoptosis, causes DNA damage, and triggers the release of pro-inflammatory cytokines. Collectively, these factors play a crucial role in the progression of cancer in individuals affected by periodontitis. Further, specific viral and bacterial agents, such as hepatitis B and C viruses, human papillomavirus (HPV), Helicobacter pylori (H. pylori), and Porphyromonas gingivalis, contribute to cancer development through distinct mechanisms. Bacterial infections have systemic implications for cancer development, while viral infections provoke immune and inflammatory responses that can lead to genetic mutations. This review will elucidate the link between periodontitis and cancers, particularly oral cancers, exploring their underlying mechanisms to provide insights for future research and treatment advancements.


Assuntos
Neoplasias Bucais , Periodontite , Humanos , Periodontite/complicações , Periodontite/microbiologia , Neoplasias Bucais/microbiologia , Neoplasias Bucais/genética , Animais , Inflamação/complicações , Porphyromonas gingivalis/patogenicidade
2.
Chem Biol Drug Des ; 103(3): e14492, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38485457

RESUMO

Recent evidence has proved that thymoquinone as a natural polyphenol has great anticancer and anti-proliferative effects in cancer cells. In this study, we aimed to examine the effects of thymoquinone on increasing cisplatin-induced apoptosis human oral squamous cell carcinoma cells and its underlying molecular mechanisms. SCC-25 cancer cells treated by thymoquinone and cisplatin with different concentrations. Cell viability will determine by using MTT assay. The concentrations of reactive oxygen species (ROS) and antioxidant activities were determined using specific related kits. DNA damage, lipid, and protein oxidation were assessed. Real-time PCR and Western blot analysis will be used to determine the expression of apoptosis-related proteins including Bax, Bcl-2, and caspase-3. Combination of thymoquinone and cisplatin suppressed synergistically SCC-25 cancer cell viability and induced apoptosis in dose-depended manner. Cell treatment with combination of thymoquinone and cisplatin led to accumulation of ROS within cells and increase in the intracellular levels of DNA damage, protein and lipid peroxidation. In addition, the combination of thymoquinone and cisplatin modulated the mRNA and protein expression levels of apoptosis-related proteins including Bax, Bcl-2, and caspase-3. Thymoquinone potentiated cisplatin anti-cancer effect on OSCC by inducing oxidative stress in cells.


Assuntos
Benzoquinonas , Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Caspase 3/genética , Caspase 3/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , Neoplasias Bucais/tratamento farmacológico , Apoptose , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Estresse Oxidativo , Linhagem Celular Tumoral
3.
Stem Cell Rev Rep ; 20(3): 688-721, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38308730

RESUMO

Biomedical engineering breakthroughs and increased patient expectations and requests for more comprehensive care are propelling the field of regenerative dentistry forward at a fast pace. Stem cells (SCs), bioactive compounds, and scaffolds are the mainstays of tissue engineering, the backbone of regenerative dentistry. Repairing damaged teeth and gums is a significant scientific problem at present. Novel therapeutic approaches for tooth and periodontal healing have been inspired by tissue engineering based on mesenchymal stem cells (MSCs). Furthermore, as a component of the MSC secretome, extracellular vesicles (EVs) have been shown to contribute to periodontal tissue repair and regeneration. The scaffold, made of an artificial extracellular matrix (ECM), acts as a supporting structure for new cell development and tissue formation. To effectively promote cell development, a scaffold must be non-toxic, biodegradable, biologically compatible, low in immunogenicity, and safe. Due to its promising biological characteristics for cell regeneration, dental tissue engineering has recently received much attention for its use of natural or synthetic polymer scaffolds with excellent mechanical properties, such as small pore size and a high surface-to-volume ratio, as a matrix. Moreover, as a bioactive material for carrying MSC-EVs, the combined application of scaffolds and MSC-EVs has a better regenerative effect on dental diseases. In this paper, we discuss how MSCs and MSC-derived EV treatment may be used to regenerate damaged teeth, and we highlight the role of various scaffolds in this process.


Assuntos
Células-Tronco Mesenquimais , Doenças Estomatognáticas , Humanos , Medicina Regenerativa , Engenharia Tecidual , Células-Tronco
4.
Chem Biol Drug Des ; 102(2): 285-291, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37060268

RESUMO

MicroRNA-34 (miR-34) is one the most important tumor suppressor miRNAs involving in the various aspects of oral cancer. The present study aimed to evaluate the effects of miR-34 restoration in OECM-1 oral cancer resistant to paclitaxel (OECM-1/PTX) and its underlying mechanisms through p53-mediated DNA damage and apoptosis. OECM-1 and OECM-1/PTX were transfected with miR-34 mimic and inhibitor. Cellular proliferation and apoptosis were evaluated through MTT assay and flow cytometry, respectively. The mRNA and protein expression levels of p53, p-glycoprotein (P-gp), ATM, ATR, CHK1, and CHK2 were assessed through qRT-PCR and western blotting. Rhodamin123 uptake assay was used to measure the P-gp activities. P53 expression was also suppressed by sing a siRNA transfection of cells. The expression levels of miR-34 were downregulated in OECM-1/PTX. Restoration of miR-34 led to increase in cytotoxic effects of paclitaxel in cells. In addition, the expression levels and activities of P-gp were reduced following miR-34 transfection. miR-34 transfection upregulated the p53, ATM, ATR, CHK1, and CHK2 expression levels in OECM-1/PTX cells. Furthermore, cells transfected with miR-34 showed higher levels of apoptosis. miR-34 restoration reverses paclitaxel resistance in OECM-1 oral cancer. The chemosensitive effects of miR-34 is mediated through increasing DNA damage and apoptosis in a p53 depended manner.


Assuntos
Carcinoma de Células Escamosas , MicroRNAs , Neoplasias Bucais , Humanos , Paclitaxel/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , MicroRNAs/genética , MicroRNAs/metabolismo , Apoptose , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Dano ao DNA , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética , Proliferação de Células , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Regulação Neoplásica da Expressão Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...