Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Nanobiotechnology ; 22(1): 171, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38610017

RESUMEN

Salivary extracellular vesicles (EVs) have emerged as key tools for non-invasive diagnostics, playing a crucial role in the early detection and monitoring of diseases. These EVs surpass whole saliva in biomarker detection due to their enhanced stability, which minimizes contamination and enzymatic degradation. The review comprehensively discusses methods for isolating, enriching, quantifying, and characterizing salivary EVs. It highlights their importance as biomarkers in oral diseases like periodontitis and oral cancer, and underscores their potential in monitoring systemic conditions. Furthermore, the review explores the therapeutic possibilities of salivary EVs, particularly in personalized medicine through engineered EVs for targeted drug delivery. The discussion also covers the current challenges and future prospects in the field, emphasizing the potential of salivary EVs in advancing clinical practice and disease management.


Asunto(s)
Vesículas Extracelulares , Neoplasias de la Boca , Humanos , Medicina de Precisión , Sistemas de Liberación de Medicamentos , Saliva
2.
J Transl Med ; 21(1): 211, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36949458

RESUMEN

The human body is colonized by abundant and diverse microorganisms, collectively known as the microbiome. The oral cavity has more than 700 species of bacteria and consists of unique microbiome niches on mucosal surfaces, on tooth hard tissue, and in saliva. The homeostatic balance between the oral microbiota and the immune system plays an indispensable role in maintaining the well-being and health status of the human host. Growing evidence has demonstrated that oral microbiota dysbiosis is actively involved in regulating the initiation and progression of an array of autoimmune diseases.Oral microbiota dysbiosis is driven by multiple factors, such as host genetic factors, dietary habits, stress, smoking, administration of antibiotics, tissue injury and infection. The dysregulation in the oral microbiome plays a crucial role in triggering and promoting autoimmune diseases via several mechanisms, including microbial translocation, molecular mimicry, autoantigen overproduction, and amplification of autoimmune responses by cytokines. Good oral hygiene behaviors, low carbohydrate diets, healthy lifestyles, usage of prebiotics, probiotics or synbiotics, oral microbiota transplantation and nanomedicine-based therapeutics are promising avenues for maintaining a balanced oral microbiome and treating oral microbiota-mediated autoimmune diseases. Thus, a comprehensive understanding of the relationship between oral microbiota dysbiosis and autoimmune diseases is critical for providing novel insights into the development of oral microbiota-based therapeutic approaches for combating these refractory diseases.


Asunto(s)
Enfermedades Autoinmunes , Microbioma Gastrointestinal , Microbiota , Probióticos , Humanos , Disbiosis/microbiología , Boca/microbiología
3.
Int J Biol Sci ; 20(1): 231-248, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38164166

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) remains a formidable clinical challenge due to its high recurrence rate and limited targeted therapeutic options. This study aims to elucidate the role of tensin 4 (TNS4) in the pathogenesis of HNSCC across clinical, cellular, and animal levels. We found a significant upregulation of TNS4 expression in HNSCC tissues compared to normal controls. Elevated levels of TNS4 were associated with adverse clinical outcomes, including diminished overall survival. Functional assays revealed that TNS4 knockdown attenuated, and its overexpression augmented, the oncogenic capabilities of HNSCC cells both in vitro and in vivo. Mechanistic studies revealed that TNS4 overexpression promotes the interaction between integrin α5 and integrin ß1, thereby activating focal adhesion kinase (FAK). This TNS4-mediated FAK activation simultaneously enhanced the PI3K/Akt signaling pathway and facilitated the interaction between TGFßRI and TGFßRII, leading to the activation of the TGFß signaling pathway. Both of these activated pathways contributed to HNSCC tumorigenesis. Additionally, we found that hypoxia-inducible factor 1α (HIF-1α) transcriptionally regulated TNS4 expression. In conclusion, our findings provide the basis for innovative TNS4-targeted therapeutic strategies, which could potentially improve prognosis and survival rates for patients with HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Proteínas Proto-Oncogénicas c-akt , Animales , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Integrina alfa5beta1 , Factor de Crecimiento Transformador beta , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Transformación Celular Neoplásica , Hipoxia , Neoplasias de Cabeza y Cuello/genética , Línea Celular Tumoral , Tensinas/metabolismo
4.
Cell Death Dis ; 15(5): 307, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693104

RESUMEN

The interplay between extracellular matrix (ECM) stiffness and the tumor microenvironment is increasingly recognized as a critical factor in cancer progression and the efficacy of immunotherapy. This review comprehensively discusses the key factors regulating ECM remodeling, including the activation of cancer-associated fibroblasts and the accumulation and crosslinking of ECM proteins. Furthermore, it provides a detailed exploration of how ECM stiffness influences the behaviors of both tumor and immune cells. Significantly, the impact of ECM stiffness on the response to various immunotherapy strategies, such as immune checkpoint blockade, adoptive cell therapy, oncolytic virus therapy, and therapeutic cancer vaccines, is thoroughly examined. The review also addresses the challenges in translating research findings into clinical practice, highlighting the need for more precise biomaterials that accurately mimic the ECM and the development of novel therapeutic strategies. The insights offered aim to guide future research, with the potential to enhance the effectiveness of cancer immunotherapy modalities.


Asunto(s)
Matriz Extracelular , Inmunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Matriz Extracelular/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales
5.
Stem Cell Res Ther ; 14(1): 55, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36978165

RESUMEN

Mesenchymal stem cell-based therapy has become an effective therapeutic approach for bone regeneration. However, there are still limitations in successful clinical translation. Recently, the secretome of mesenchymal stem cells, especially exosome, plays a critical role in promoting bone repair and regeneration. Exosomes are nanosized, lipid bilayer-enclosed structures carrying proteins, lipids, RNAs, metabolites, growth factors, and cytokines and have attracted great attention for their potential application in bone regenerative medicine. In addition, preconditioning of parental cells and exosome engineering can enhance the regenerative potential of exosomes for treating bone defects. Moreover, with recent advancements in various biomaterials to enhance the therapeutic functions of exosomes, biomaterial-assisted exosomes have become a promising strategy for bone regeneration. This review discusses different insights regarding the roles of exosomes in bone regeneration and summarizes the applications of engineering exosomes and biomaterial-assisted exosomes as safe and versatile bone regeneration agent delivery platforms. The current hurdles of transitioning exosomes from bench to bedside are also discussed.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Materiales Biocompatibles/uso terapéutico , Exosomas/genética , Exosomas/metabolismo , Regeneración Ósea/genética , Medicina Regenerativa , Células Madre Mesenquimatosas/metabolismo
6.
Front Biosci (Landmark Ed) ; 28(5): 85, 2023 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-37258472

RESUMEN

BACKGROUND: Human dental stem cells (DSCs) are excellent sources of cells for treating dental and craniofacial diseases. However, the mechanisms regulating DSC osteogenic differentiation are still unclear. In this study, we aimed to determine the role of Krüppel-like factor 9 (KLF9) in regulating the biological functions of DSCs and explore the underlying molecular mechanisms. METHODS: Bioinformatic analyses, quantitative real-time polymerase chain reaction (qRT‒PCR) and Western blotting were performed to determine the KLF9 level during osteogenic differentiation of DSCs. The effects of KLF9 depletion or overexpression on DSC osteogenic differentiation were then evaluated. The osteogenic potential and associated mineralized nodule-forming activities of DSCs were monitored via Alizarin red S staining and quantitative analyses of osteogenic markers. The regulatory effect of KLF9 on the Notch1 signaling pathway was analyzed by luciferase reporter assays. RESULTS: KLF9 mRNA expression was consistently increased during mesenchymal stem cell osteogenic differentiation in multiple public datasets, and our qRT‒PCR and Western blotting data further validated this finding. In addition, KLF9 depletion promoted proliferation and suppressed osteogenic differentiation of DSCs, while enforced expression of KLF9 promoted the DSC osteogenic potential. Mechanistically, KLF9 negatively regulated the Notch1-mediated signaling pathway by directly binding to the Notch1 promoter. More importantly, Notch1 inhibition/overexpression partially rescued the suppressive/enhancing effects of KLF9 depletion/overexpression on the osteogenic differentiation of DSCs, indicating that Notch1 is a functional downstream target of KLF9. CONCLUSIONS: In summary, our results strongly demonstrate that KLF9 is a crucial transcription factor that controls the osteogenic differentiation of DSCs by negatively regulating the Notch1 signaling pathway.


Asunto(s)
MicroARNs , Osteogénesis , Humanos , Osteogénesis/genética , Células Madre/metabolismo , Diferenciación Celular/genética , Transducción de Señal , Regulación de la Expresión Génica , Células Cultivadas , MicroARNs/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo
7.
Cancers (Basel) ; 15(23)2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-38067199

RESUMEN

BACKGROUD: The stratification of head and neck squamous cell carcinoma (HNSCC) patients based on prognostic differences is critical for therapeutic guidance. This study was designed to construct a predictive signature derived from T-cell receptor-related genes (TCRRGs) to forecast the clinical outcomes in HNSCC. METHODS: We sourced gene expression profiles from The Cancer Genome Atlas (TCGA) HNSCC dataset, GSE41613, and GSE65858 datasets. Utilizing consensus clustering analysis, we identified two distinct HNSCC clusters according to TCRRG expression. A TCRRG-based signature was subsequently developed and validated across diverse independent HNSCC cohorts. Moreover, we established a nomogram model based on TCRRGs. We further explored differences in immune landscapes between high- and low-risk groups. RESULTS: The TCGA HNSCC dataset was stratified into two clusters, displaying marked variations in both overall survival (OS) and immune cell infiltration. Furthermore, we developed a robust prognostic signature based on TCRRG utilizing the TCGA HNSCC train cohort, and its prognostic efficacy was validated in the TCGA HNSCC test cohort, GSE41613, and GSE65858. Importantly, the high-risk group was characterized by a suppressive immune microenvironment, in contrast to the low-risk group. Our study successfully developed a robust TCRRG-based signature that accurately predicts clinical outcomes in HNSCC, offering valuable strategies for improved treatments.

8.
Front Oncol ; 11: 770241, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35127477

RESUMEN

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is still a menace to public wellbeing globally. However, the underlying molecular events influencing the carcinogenesis and prognosis of HNSCC are poorly known. METHODS: Gene expression profiles of The Cancer Genome Atlas (TCGA) HNSCC dataset and GSE37991 were downloaded from the TCGA database and gene expression omnibus, respectively. The common differentially expressed metabolic enzymes (DEMEs) between HNSCC tissues and normal controls were screened out. Then a DEME-based molecular signature and a clinically practical nomogram model were constructed and validated. RESULTS: A total of 23 commonly upregulated and 9 commonly downregulated DEMEs were identified in TCGA HNSCC and GSE37991. Gene ontology analyses of the common DEMEs revealed that alpha-amino acid metabolic process, glycosyl compound metabolic process, and cellular amino acid metabolic process were enriched. Based on the TCGA HNSCC cohort, we have built up a robust DEME-based prognostic signature including HPRT1, PLOD2, ASNS, TXNRD1, CYP27B1, and FUT6 for predicting the clinical outcome of HNSCC. Furthermore, this prognosis signature was successfully validated in another independent cohort GSE65858. Moreover, a potent prognostic signature-based nomogram model was constructed to provide personalized therapeutic guidance for treating HNSCC. In vitro experiment revealed that the knockdown of TXNRD1 suppressed malignant activities of HNSCC cells. CONCLUSION: Our study has successfully developed a robust DEME-based signature for predicting the prognosis of HNSCC. Moreover, the nomogram model might provide useful guidance for the precision treatment of HNSCC.

9.
Front Genet ; 12: 750990, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34764982

RESUMEN

Mesenchymal stem cells (MSCs) are promising seed cells in tissue repair and regeneration due to their featured properties of self-renewal and multipotency. However, a growing body of evidence has demonstrated that MSCs exert biological functions mainly through secreting exosomes. Exosomes, which contain RNA, proteins, lipids, and metabolites, are new players in regulating many fundamental processes and play important roles in regenerative medicine. Exosomes not only mimic the effects of their parent cells but also possess many advantages such as high drug loading capacity, low immunogenicity, excellent biocompatibility, and low side effects. Currently, a total of 6 different dental stem cells (DSCs) including dental pulp stem cells (DPSCs), stem cells from exfoliated deciduous teeth (SHEDs), periodontal ligament stem cells (PDLSCs), dental follicle progenitor cells (DFPCs), stem cells from apical papilla (SCAPs) and gingival mesenchymal stem cells (GMSCs) have been isolated and identified. DSC-derived exosomes (DSC-Exos) are actively involved in intercellular communication, anti-inflammation, osteogenesis, angiogenesis, immunomodulation, nurturing neurons, and promoting tumor cell apoptosis. In this review, we will critically review the emerging role and clinical application potential of DSC-Exos.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA