Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Dent Educ ; 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138879

RESUMEN

PURPOSE: Altering the curriculum of a program can have negative repercussions for the student experience, including peer mentorships and interclass relationships. This study investigated the effect of curriculum reform on students' emotional and social well-being in a predoctoral dental program. We explored if any of these consequences could be related to stereotype threat. METHODS: We utilized a quasi-experimental design with two different treatments, New Curriculum Treatment (New-CT, n = 44) and Past Curriculum Treatment (Past-CT, n = 43). Quantitative data were collected through surveys to assess students' perceptions of curriculum changes and their impacts on anxiety, confidence, and clinical performance. Qualitative data were gathered via semi-structured interviews to explore personal experiences of stereotype threat and its implications on peer relationships and mentorship dynamics. RESULTS: The findings suggest significant effects of curriculum changes on interpersonal relationships. Past-CT viewed New-CT as overconfident, while New-CT felt heightened performance pressure. Thematic and interview analyses revealed deep-rooted tensions, with New-CT feeling mistrusted and Past-CT resenting New-CT's perceived accelerated competence. Stereotype threat was identified as a key factor worsening these inter-group tensions and affecting clinical performance and relationships. CONCLUSIONS: Curriculum changes in dental education can significantly affect students' well-being, with stereotype threat playing a critical role in these dynamics. When making changes to the structure, sequencing, or content of a program, administrators need to be aware of the potential ramifications these changes could have on students' relationships with their peers.

2.
J Clin Pediatr Dent ; 48(4): 26-37, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39087211

RESUMEN

Poor oral health during childhood can lead to various oral diseases and have long-term implications for dental health. Innovative and engaging oral health educational approaches such as game-based teaching have emerged as a promising modality for health education. This systematic review examined the effectiveness of game-based teaching methods on the oral health of children (4-12 yrs). Scopus, Medline and Web of Science databases were searched according to specific inclusion and exclusion criteria. Inclusion criteria included randomised trials that compared traditional methods of oral health education with game-based interventions in preschoolers and school-age children. The quality of the data was determined using Cochrane risk-of-bias tool for randomized trials (ROB-2). A total of seven studies that examined 1097 children (4-12 yrs) were included in this systematic review with the association of game-based teaching of oral health. The findings indicated that the utilization of game-based methods significantly improved children's oral health outcomes when compared to traditional teaching approaches. Specifically, the game-based interventions demonstrated positive effects on various aspects of oral health, including enhanced oral health knowledge, improved oral hygiene scores, and reductions in debris and plaque scores. The game-based interventions were found to be more effective in promoting oral health when compared to conventional methods of teaching, such as verbal instructions or educational posters. Based on the limited evidence available, game-based teaching appears to be an effective approach for promoting oral health among children, consistently demonstrating positive outcomes, including improved oral health knowledge, enhanced oral hygiene scores, and reductions in debris and plaque scores. Further well-designed trials adhering to reporting guidelines and using objective measures are necessary before outlining universal guidelines for best practice.


Asunto(s)
Educación en Salud Dental , Salud Bucal , Ensayos Clínicos Controlados Aleatorios como Asunto , Humanos , Niño , Salud Bucal/educación , Educación en Salud Dental/métodos , Preescolar , Higiene Bucal/educación
3.
Front Oncol ; 14: 1383062, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38915370

RESUMEN

This review presents an in-depth analysis of the immense potential of CRISPR-Cas9 technology in revolutionizing oral cancer research. It underscores the inherent limitations of conventional treatments while emphasizing the pressing need for groundbreaking approaches. The unparalleled capability of CRISPR-Cas9 to precisely target and modify specific genes involved in cancer progression heralds a new era in therapeutic intervention. Employing genome-wide CRISPR screens, vulnerabilities in oral cancer cells can be identified, thereby unravelling promising targets for therapeutic interventions. In the realm of oral cancer, the disruptive power of CRISPR-Cas9 manifests through its capacity to perturb genes that are intricately associated with drug resistance, consequently augmenting the efficacy of chemotherapy. To address the challenges that arise, this review diligently examines pertinent issues such as off-target effects, efficient delivery mechanisms, and the ethical considerations surrounding germline editing. Through precise gene editing, facilitated by CRISPR/Cas9, it becomes possible to overcome drug resistance by rectifying mutations, thereby enhancing the efficacy of personalized treatment strategies. This review delves into the prospects of CRISPR-Cas9, illuminating its potential applications in the domains of medicine, agriculture, and biotechnology. It is paramount to emphasize the necessity of ongoing research endeavors and the imperative to develop targeted therapies tailored specifically for oral cancer. By embracing this comprehensive overview, we can pave the way for ground-breaking treatments that instill renewed hope for enhanced outcomes in individuals afflicted by oral cancer.

5.
Phytomedicine ; 129: 155511, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38723523

RESUMEN

BACKGROUND: Mitochondrial dysfunction associated with mitochondrial DNA mutations, enzyme defects, generation of ROS, and altered oxidative homeostasis is known to induce oral carcinogenesis during exposure to arecoline. Butein, a natural small molecule from Butea monosperma, possesses anti-inflammatory, anti-diabetic, and anti-cancer effects. However, the role of butein in the mitochondrial quality control mechanism has not been illuminated clearly. PURPOSE: This study aimed to explore the role of butein in preserving mitochondrial quality control during arecoline-induced mitochondrial dysfunction in oral cancer to curtail the early onset of carcinogenesis. METHODS: Cell viability was evaluated by MTT assay. The relative protein expressions were determined by western blotting. Immunofluorescence and confocal imaging were used to analyze the relative fluorescence and co-localization of proteins. Respective siRNAs were used to examine the knockdown-based studies. RESULTS: Butein, in the presence of arecoline, significantly caused a decrease in mitochondrial hyperpolarization and ROS levels in oral cancer cells. Mechanistically, we found an increase in COXIV, TOM20, and PGC1α expression during butein treatment, and inhibition of PGC1α blunted mitochondrial biogenesis and decreased the mitochondrial pool. Moreover, the fission protein MTP18, and its molecular partners DRP1 and MFF were dose-dependently increased during butein treatment to maintain mitochondria mass. In addition, we also found increased expression of various mitophagy proteins, including PINK1, Parkin, and LC3 during butein treatment, suggesting the clearance of damaged mitochondria to maintain a healthy mitochondrial pool. Interestingly, butein increased the activity of SIRT1 to enhance the functional mitochondrial pool, and inhibition of SIRT1 found to reduce the mitochondrial levels, as evident from the decrease in the expression of PGC1α and MTP18 in oral cancer cells. CONCLUSION: Our study proved that SIRT1 maintains a functional mitochondrial pool through PGC1α and MTP18 for biogenesis and fission of mitochondria during arecoline exposure and could decrease the risk of mitochondria dysfunctionality associated with the onset of oral carcinogenesis.


Asunto(s)
Arecolina , Chalconas , Mitocondrias , Neoplasias de la Boca , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Especies Reactivas de Oxígeno , Sirtuina 1 , Humanos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Arecolina/farmacología , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Chalconas/farmacología , Sirtuina 1/metabolismo , Supervivencia Celular/efectos de los fármacos
7.
Cancer Lett ; 590: 216843, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38579893

RESUMEN

Recurrent chemotherapy-induced senescence and resistance are attributed to the polyploidization of cancer cells that involve genomic instability and poor prognosis due to their unique form of cellular plasticity. Autophagy, a pre-dominant cell survival mechanism, is crucial during carcinogenesis and chemotherapeutic stress, favouring polyploidization. The selective autophagic degradation of essential proteins associated with cell cycle progression checkpoints deregulate mitosis fidelity and genomic integrity, imparting polyploidization of cancer cells. In connection with cytokinesis failure and endoreduplication, autophagy promotes the formation, maintenance, and generation of the progeny of polyploid giant cancer cells. The polyploid cancer cells embark on autophagy-guarded elevation in the expression of stem cell markers, along with triggered epithelial and mesenchymal transition and senescence. The senescent polyploid escapers represent a high autophagic index than the polyploid progeny, suggesting regaining autophagy induction and subsequent autophagic degradation, which is essential for escaping from senescence/polyploidy, leading to a higher proliferative phenotypic progeny. This review documents the various causes of polyploidy and its consequences in cancer with relevance to autophagy modulation and its targeting for therapeutic intervention as a novel therapeutic strategy for personalized and precision medicine.


Asunto(s)
Autofagia , Senescencia Celular , Neoplasias , Células Madre Neoplásicas , Poliploidía , Humanos , Senescencia Celular/efectos de los fármacos , Neoplasias/patología , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Animales , Transición Epitelial-Mesenquimal
8.
Autophagy ; 20(6): 1359-1382, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38447939

RESUMEN

Mitophagy involves the selective elimination of defective mitochondria during chemotherapeutic stress to maintain mitochondrial homeostasis and sustain cancer growth. Here, we showed that CLU (clusterin) is localized to mitochondria to induce mitophagy controlling mitochondrial damage in oral cancer cells. Moreover, overexpression and knockdown of CLU establish its mitophagy-specific role, where CLU acts as an adaptor protein that coordinately interacts with BAX and LC3 recruiting autophagic machinery around damaged mitochondria in response to cisplatin treatment. Interestingly, CLU triggers class III phosphatidylinositol 3-kinase (PtdIns3K) activity around damaged mitochondria, and inhibition of mitophagic flux causes the accumulation of excessive mitophagosomes resulting in reactive oxygen species (ROS)-dependent apoptosis during cisplatin treatment in oral cancer cells. In parallel, we determined that PPARGC1A/PGC1α (PPARG coactivator 1 alpha) activates mitochondrial biogenesis during CLU-induced mitophagy to maintain the mitochondrial pool. Intriguingly, PPARGC1A inhibition through small interfering RNA (siPPARGC1A) and pharmacological inhibitor (SR-18292) treatment counteracts CLU-dependent cytoprotection leading to mitophagy-associated cell death. Furthermore, co-treatment of SR-18292 with cisplatin synergistically suppresses tumor growth in oral cancer xenograft models. In conclusion, CLU and PPARGC1A are essential for sustained cancer cell growth by activating mitophagy and mitochondrial biogenesis, respectively, and their inhibition could provide better therapeutic benefits against oral cancer.


Asunto(s)
Supervivencia Celular , Clusterina , Mitocondrias , Mitofagia , Neoplasias de la Boca , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Humanos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Clusterina/metabolismo , Clusterina/genética , Mitofagia/efectos de los fármacos , Mitofagia/fisiología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/genética , Animales , Supervivencia Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Biogénesis de Organelos , Ratones , Apoptosis/efectos de los fármacos , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Autofagia/fisiología , Autofagia/efectos de los fármacos
9.
BMC Oral Health ; 24(1): 94, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38229101

RESUMEN

Monocytes and their macrophage progeny are thought to be involved in tissue and alveolar bone destruction in periodontal disease. It has been documented that the proportion of (CD14 + CD16+) non-classical monocytes in the blood are elevated in chronic periodontitis;A total of 20 chronic generalized periodontitis patients who were otherwise healthy, were recruited for this study. At baseline and 3 weeks after non-surgical periodontal treatment, peripheral blood was obtained to assess the levels of C-reactive protein (CRP) and the proportion of monocyte subsets. Monocyte subsets were assessed using flow cytometry;The mean percentage of CD14 + CD16+ non-classical monocytes in the peripheral blood sample at baseline was 13.95 + 2.09, that reduced to 8.94 + 1.23 3 weeks after non-surgical treatment. A distinct significant reduction in the percentage of non-classical monocytes and a concomitant increase in classical monocytes were observed following periodontal treatment compared to baseline. There was a significant reduction in the all the periodontal parameters and CRP levels 3 weeks post non-surgical periodontal treatment. A positive correlation between CRP and percentage of non-classical monocytes was also observed; Periodontal treatment potentially modulates the host response effectively.


Asunto(s)
Periodontitis Crónica , Monocitos , Humanos , Monocitos/metabolismo , Receptores de IgG/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Macrófagos , Periodontitis Crónica/terapia , Periodontitis Crónica/metabolismo
10.
J Am Dent Assoc ; 155(3): 191-192, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38244018
11.
Front Oncol ; 13: 1247399, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38170015

RESUMEN

The clinical management of oral cancer is often frequented with challenges that arise from relapse, recurrence, invasion and resistance towards the cornerstone chemo and radiation therapies. The recent conceptual advancement in oncology has substantiated the role of cancer stem cells (CSC) as a predominant player of these intricacies. CSC are a sub-group of tumor population with inherent adroitness to self-renew with high plasticity. During tumor evolution, the structural and functional reprogramming persuades the cancer cells to acquire stem-cell like properties, thus presenting them with higher survival abilities and treatment resistance. An appraisal on key features that govern the stemness is of prime importance to confront the current challenges encountered in oral cancer. The nurturing niche of CSC for maintaining its stemness characteristics is thought to be modulated by complex multi-layered components encompassing neoplastic cells, extracellular matrix, acellular components, circulatory vessels, various cascading signaling molecules and stromal cells. This review focuses on recapitulating both intrinsic and extrinsic mechanisms that impart the stemness. There are contemplating evidences that demonstrate the role of transcription factors (TF) in sustaining the neoplastic stem cell's pluripotency and plasticity alongside the miRNA in regulation of crucial genes involved in the transformation of normal oral mucosa to malignancy. This review illustrates the interplay between miRNA and various known TF of oral cancer such as c-Myc, SOX, STAT, NANOG and OCT in orchestrating the stemness and resistance features. Further, the cross-talks involved in tumor micro-environment inclusive of cytokines, macrophages, extra cellular matrix, angiogenesis leading pathways and influential factors of hypoxia on tumorigenesis and CSC survival have been elucidated. Finally, external factorial influence of oral microbiome gained due to the dysbiosis is also emphasized. There are growing confirmations of the possible roles of microbiomes in the progression of oral cancer. Given this, an attempt has been made to explore the potential links including EMT and signaling pathways towards resistance and stemness. This review provides a spectrum of understanding on stemness and progression of oral cancers at various regulatory levels along with their current therapeutic knowledge. These mechanisms could be exploited for future research to expand potential treatment strategies.

12.
Front Oncol ; 13: 1183766, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38234400

RESUMEN

Oral cancer is one of the 19most rapidly progressing cancers associated with significant mortality, owing to its extreme degree of invasiveness and aggressive inclination. The early occurrences of this cancer can be clinically deceiving leading to a poor overall survival rate. The primary concerns from a clinical perspective include delayed diagnosis, rapid disease progression, resistance to various chemotherapeutic regimens, and aggressive metastasis, which collectively pose a substantial threat to prognosis. Conventional clinical practices observed since antiquity no longer offer the best possible options to circumvent these roadblocks. The world of current cancer research has been revolutionized with the advent of state-of-the-art technology-driven strategies that offer a ray of hope in confronting said challenges by highlighting the crucial underlying molecular mechanisms and drivers. In recent years, bioinformatics and Machine Learning (ML) techniques have enhanced the possibility of early detection, evaluation of prognosis, and individualization of therapy. This review elaborates on the application of the aforesaid techniques in unraveling potential hints from omics big data to address the complexities existing in various clinical facets of oral cancer. The first section demonstrates the utilization of omics data and ML to disentangle the impediments related to diagnosis. This includes the application of technology-based strategies to optimize early detection, classification, and staging via uncovering biomarkers and molecular signatures. Furthermore, breakthrough concepts such as salivaomics-driven non-invasive biomarker discovery and omics-complemented surgical interventions are articulated in detail. In the following part, the identification of novel disease-specific targets alongside potential therapeutic agents to confront oral cancer via omics-based methodologies is presented. Additionally, a special emphasis is placed on drug resistance, precision medicine, and drug repurposing. In the final section, we discuss the research approaches oriented toward unveiling the prognostic biomarkers and constructing prediction models to capture the metastatic potential of the tumors. Overall, we intend to provide a bird's eye view of the various omics, bioinformatics, and ML approaches currently being used in oral cancer research through relevant case studies.

14.
J. appl. oral sci ; 29: e20200751, 2021. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1154613

RESUMEN

Abstract Objective To investigate the Ki 67 expression and its correlation with clinicopathological features and 3 years as well as 5 years survival rate in oral squamous cell carcinoma (OSCC). Methodology Total 217cases of OSCC primarily treated with surgery with or without radiation were included. All patients were followed up for 3 years and 150 were followed up of 5 years for disease free survival. The immunohistochemistry was carried out on neutral buffered formalin fixed paraffin embedded tissue to evaluate the expression of Ki67. Results The Ki67 labeling index (LI) was significantly higher with respect to adverse clinicopathological parameters such as histopathological grading (p<0.001), clinical TNM staging (p<0.001) and nodal metastasis (p<0.001). The OSCC patients survived for less than 3 and 5 years were showed significantly higher Ki67 LI as compared to diseases free survived more than 3 and 5 years(p<0.001). The three years survival rate of OSCC patient significantly higher with low Ki67 LI (≤45) 96.2%, followed by moderate Ki67 LI (46 to 60) 60.7% and high Ki67 LI (≥61) 37.7% (p<0.001). The five years survival rate of OSCC patient statistically significantly higher with low Ki67 LI (≤45)93.3%, followed by moderate Ki67 LI (46 to 60) 46.8% and Ki67 LI (≥61) 23.3% (p<0.001). Conclusion The measurement of cell proliferative activity by using Ki67 antigen expression in individual OSCC might provide unique, predictive information on clinical outcome, prognosis and deciding treatment modalities in OSCC.


Asunto(s)
Humanos , Neoplasias de la Boca , Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Pronóstico , Biomarcadores de Tumor , Antígeno Ki-67 , Carcinoma de Células Escamosas de Cabeza y Cuello
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA