Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
J Cell Mol Med ; 28(18): e70112, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39320020

RESUMEN

Oral submucous fibrosis (OSF) is a precancerous condition in the oral cavity, which is closely related to the myofibroblast conversion of buccal mucosal fibroblasts (BMFs) after chronic consumption of areca nut. Emerging evidence suggests pyroptosis, a form of programmed cell death that is mediated by inflammasome, is implicated in persistent myofibroblast activation and fibrosis. Besides, numerous studies have demonstrated the effects of non-coding RNAs on pyroptosis and myofibroblast activities. Herein, we aimed to target key long non-coding RNA PVT1 with natural compound, carvacrol, to alleviate pyroptosis and myofibroblast activation in OSF. We first identified PVT1 was downregulated in the carvacrol-treated fBMFs and then demonstrated that myofibroblast features and expression of pyroptosis makers were all reduced in response to carvacrol treatment. Subsequently, we analysed the expression of PVT1 and found that PVT1 was aberrantly upregulated in OSF specimens and positively correlated with several fibrosis markers. After revealing the suppressive effects of carvacrol on myofibroblast characterisitcs and pyroptosis were mediated by repression of PVT1, we then explored the potential mechanisms. Our data showed that PVT1 may serve as a sponge of microRNA(miR)-20a to mitigate the myofibroblast activation and pyroptosis. Altogether, these findings indicated that the anti-fibrosis effects of carvacrol merit consideration and may be due to the attenuation of pyroptosis and myofibroblast activation by targeting the PVT1/miR-20a axis.


Asunto(s)
Cimenos , MicroARNs , Miofibroblastos , Fibrosis de la Submucosa Bucal , Piroptosis , ARN Largo no Codificante , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Piroptosis/efectos de los fármacos , Piroptosis/genética , MicroARNs/genética , MicroARNs/metabolismo , Humanos , Cimenos/farmacología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Progresión de la Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos
2.
Cytokine ; 183: 156742, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39217916

RESUMEN

OBJECTIVES: The M1/M2 macrophage framework is crucial in organ fibrosis and its progression to malignancy. This study investigated the possible role of M1/M2 macrophage interplay in the pathogenesis of oral submucous fibrosis (OSF) and its malignant transformation by analysing immunohistochemical expression of CD11c (M1) and CD163 (M2) markers. METHODS: Immunohistochemistry was performed using primary antibodies against CD11c and CD163 on ten formalin-fixed paraffin-embedded tissue blocks for each group: (i) Stage 1 OSF, (ii) Stage 2 OSF, (iii) Stage 3 OSF, (iv) Stage 4 OSF, (v) well-differentiated squamous cell carcinoma (WDSCC) with OSF, and (vi) WDSCC without OSF. Ten cases of healthy buccal mucosa (NOM) served as controls. RESULTS: Epithelial quick scores of M1 (CD11c) in NOM, Stages 1-4 OSF, and WDSCC with and without OSF were 0, 1.8, 2.9, 0.4, 0, 0, and 0, while connective tissue scores were 0, 3.2, 4.3, 2.7, 0.5, 1.2, and 2.4, respectively. Epithelial scores for M2 (CD163) were 0, 0.8, 0.8, 2.1, 0.6, 0.8, and 0.2, and connective tissue scores were 0, 1.8, 2.6, 3.9, 2.2, 5, and 4.4, respectively. Stages 3 and 4 OSF, WDSCC with and without OSF exhibited higher M2/M1 ratios compared to NOM and Stages 1-2 OSF. CONCLUSION: The interaction between M1 (CD11c) and M2 (CD163) macrophages, leading to M2 polarisation, plays a crucial role in the pathogenesis of OSF and its potential malignant transformation.


Asunto(s)
Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Antígeno CD11c , Transformación Celular Neoplásica , Inmunohistoquímica , Fibrosis de la Submucosa Bucal , Receptores de Superficie Celular , Humanos , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/metabolismo , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Receptores de Superficie Celular/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígeno CD11c/metabolismo , Antígenos CD/metabolismo , Masculino , Femenino , Macrófagos/metabolismo , Macrófagos/patología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Persona de Mediana Edad , Adulto , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Mucosa Bucal/patología , Mucosa Bucal/metabolismo
3.
Clin Biochem ; 130: 110790, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38969054

RESUMEN

This study aims to investigate the alteration of salivary biomarker profiling in the development of oral submucous fibrosis (OSMF) and to explore the influence of saliva in the diagnosis of OSMF. A systematic search of published articles using the PRISMA guidelines was conducted to identify relevant studies on OSMF and saliva. All eligible studies, including case-control, cross-sectional studies, cohort, and pilot studies, contained the evaluation of salivary biomarker profiling in patients with OSMF. Salivary biomarker data from 28 selected articles were categorized into nine groups, and their mean values were determined. A three-step meta-analysis was performed by grouping salivary biomarker profiling into more heterogeneous categories based on OSMF classification, considering functional, histological, and clinical grading. The salivary biomarker profiling analysis revealed significant alterations in all markers, indicating their efficacy in OSMF diagnosis. Subgroup analyses highlighted significant associations in oxidative stress and protein with increased mean values, particularly emphasizing lipid peroxidase (LPO), malondialdehyde (MDA), and lactate dehydrogenase (LDH). Conversely, decreased mean values were observed in glutathione, glutathione peroxidase (GPx), superoxide dismutase (SOD), and vitamins. Notably, OSMF grading analysis demonstrated a significant difference in weighted effect sizes for histological grading, particularly in stage IV. The study underscores the alteration of specific salivary biomarkers, particularly those associated with LPO, MDA, LDH, glutathione, GPx, SOD, and vitamins, in diagnosing and grading OSMF.


Asunto(s)
Biomarcadores , Glutatión Peroxidasa , Malondialdehído , Fibrosis de la Submucosa Bucal , Saliva , Superóxido Dismutasa , Humanos , Biomarcadores/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Malondialdehído/metabolismo , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/diagnóstico , Estrés Oxidativo , Saliva/metabolismo , Superóxido Dismutasa/metabolismo , Vitaminas
4.
J Cancer Res Ther ; 20(3): 788-792, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-39023584

RESUMEN

BACKGROUND AND AIM: Precancer biomarkers help in early detection and management of oral potentially malignant disorders (OPMDs). Interleukin-1ß (IL-1ß), a biomarker, is known to be altered in oral submucous fibrosis (OSMF) and oral leukoplakia (OL). Therefore, we evaluated and compared the serum and salivary IL-1ß levels in patients with OSMF/oral leukoplakia and in gender- and age-matched healthy individuals. MATERIALS AND METHODS: An in vivo, prospective, observational study was conducted on 40 subjects. Subjects were divided into two groups with 20 individuals in each group, that is, Group I: OSMF/oral leukoplakia and Group II: control group. Salivary and serum IL-1ß levels were quantitatively estimated using enzyme-linked immunosorbent assay (ELISA). The statistical tests used were unpaired t-test and Chi-square test. RESULTS: The serum IL-1ß levels were significantly (P 0.001) lesser in Group I in comparison to Group II. The salivary IL-1ß levels remained insignificant between both the groups. However, in both the groups, the salivary IL-1ß levels were significantly higher compared to the serum IL-1ß levels. CONCLUSION: We found that the serum IL-1ß level can be considered as a prospective biomarker for dysplasia, whereas salivary IL-1ß alone needs more elaborated studies to account for its application as a potential biomarker in OPMD.


Asunto(s)
Interleucina-1beta , Leucoplasia Bucal , Neoplasias de la Boca , Fibrosis de la Submucosa Bucal , Lesiones Precancerosas , Saliva , Humanos , Interleucina-1beta/sangre , Interleucina-1beta/análisis , Interleucina-1beta/metabolismo , Masculino , Femenino , Saliva/metabolismo , Saliva/química , Leucoplasia Bucal/sangre , Leucoplasia Bucal/diagnóstico , Leucoplasia Bucal/metabolismo , Leucoplasia Bucal/patología , Estudios Prospectivos , Adulto , Persona de Mediana Edad , Lesiones Precancerosas/sangre , Lesiones Precancerosas/diagnóstico , Lesiones Precancerosas/patología , Lesiones Precancerosas/metabolismo , Fibrosis de la Submucosa Bucal/sangre , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/diagnóstico , Fibrosis de la Submucosa Bucal/patología , Neoplasias de la Boca/sangre , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Estudios de Casos y Controles , Biomarcadores/sangre , Biomarcadores/análisis
5.
Arch Oral Biol ; 167: 106052, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39047597

RESUMEN

OBJECTIVE: To determine the biological effects of arecoline on oral submucous fibrosis (OSF). DESIGN: The differential genes between OSF tissue and normal oral tissue were collected form GSE64216 dataset, analyzed by Gene Expression Omnibus (GEO) database. Real-time PCR and immunohistochemistry were used to analyze the expression of IL-4 gene and protein in oral tissue. Enzyme-linked immunosorbent assay (ELISA) was used to analyze the expression of exocrine IL-4 protein in human oral fibroblasts (HOF) pre-treated by arecoline. Cell Counting Kit-8 (CCK-8) and transwell assays were used to analyze the proliferation and migration of HOF cells, respectively. After IL-4 was knocked down by short hairpin (sh) plasmid, the proliferation and migration of HOF cells were detected. Flow cytometry was used to analyze the proportion of M2-macrophages. Real-time PCR and immunohistochemistry were used to verify the expression of biomarker proteins of macrophages in OSF tissues. RESULTS: The expression of IL-4 gene and protein were both up-regulated in OSF tissue. Arecoline could enhance the expression of IL-4 gene and exocrine protein in HOF cells, and promote the proliferation and migration of HOF cells. While knockdown of IL-4 could inhibit arecoline-induced proliferation and migration in HOF cells. The results of flow cytometry showed that recombinant human IL-4 (rhIL-4) protein could increase the proportion of M2-macrophages. Similarly, the results of real-time PCR and immunohistochemistry showed the expression of ARG1 (Biomarker proteins of M2-macrophage) was up-regulated in OSF tissues. CONCLUSION: Arecoline promotes activation of fibroblasts and polarization of M2-macrophages by up-regulating the expression of IL-4.


Asunto(s)
Arecolina , Movimiento Celular , Proliferación Celular , Fibroblastos , Interleucina-4 , Macrófagos , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba , Interleucina-4/farmacología , Humanos , Arecolina/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proliferación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Movimiento Celular/efectos de los fármacos , Fibrosis de la Submucosa Bucal/metabolismo , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Citometría de Flujo , Células Cultivadas
6.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(4): 444-451, 2024 Aug 01.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-39049631

RESUMEN

OBJECTIVES: This study aims to investigate the primary target and potential mechanism of mangiferin (MF) in treating oral submucous fibrosis (OSF) through Gene Expression Omnibus (GEO) database chip mining, network pharmacology, and molecular docking techniques. METHODS: Potential therapeutic targets for OSF were identified using GEO chip data. The potential targets of MF were predicted, and disease-related targets for OSF were collected from databases. A Venn diagram was created using the EVenn platform to identify overlapping targets. The protein-protein interaction (PPI) network was constructed using the STRING database. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed using the DAVID platform. Cytoscape 3.10.1 software was used to visualize a drug-target-pathway-disease network, while AutoDocktools 1.5.6 software was employed for molecular docking analysis. RESULTS: A total of 356 potential targets for MF and 360 disease-related targets for OSF were obtained from multiple databases. The top 15 key target proteins in the PPI network were selected as significant candidates. GO function and KEGG pathway enrichment analyses revealed that MF treatment primarily involved advanced glycation end products-receptor (AGE-RAGE), epidermal growth factor receptor (EGFR), and other signaling pathways associated with OSF pathogenesis. Molecular docking analysis demonstrated that MF exhibited a strong binding activity toward AKT serine kinase 1 (AKT1), tumor necrosis factor (TNF), and other core targets. CONCLUSIONS: These findings suggest that MF may exert its therapeutic effects on OSF through a multitarget approach involving various signaling pathways.


Asunto(s)
Simulación del Acoplamiento Molecular , Farmacología en Red , Fibrosis de la Submucosa Bucal , Mapas de Interacción de Proteínas , Xantonas , Xantonas/uso terapéutico , Xantonas/farmacología , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Fibrosis de la Submucosa Bucal/metabolismo , Humanos , Ontología de Genes , Minería de Datos , Receptores ErbB/metabolismo , Programas Informáticos , Transducción de Señal
7.
J Tradit Chin Med ; 44(4): 688-693, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39066529

RESUMEN

OBJECTIVE: To investigate the protective effect of the Chinese herbal formula of Jiedu Huayu decoction (, JHD) on oral mucosa of rats with oral submucosal fibrosis (OSF) and its potential mechanism of action. METHODS: Sprague-Dawley male OSF model rats were constructed by injection of betaine and topical rubbing and were randomly grouped and administered by gavage for 4 weeks. Mouth opening and buccal mucosa scores interleukin levels and the expression of Axin and ß-catenin proteins or genes were measured before and after drug administration. RESULTS: After treatment with JHD the buccal mucosal lesions of rats were significantly reduced Axin protein and mRNA expression were significantly increased ß-catenin protein and mRNA expression were significantly decreased interleukin-1ß and interleukin-6 levels were decreased and interleukin-10 levels were increased. CONCLUSION: The mechanism of action of JHD can effectively alleviate the pathological damage of buccal mucosa in OSF rats which may be related to the promotion of Axin expression and inhibition of ß-catenin expression.


Asunto(s)
Proteína Axina , Medicamentos Herbarios Chinos , Mucosa Bucal , Ratas Sprague-Dawley , beta Catenina , Animales , Medicamentos Herbarios Chinos/administración & dosificación , Masculino , beta Catenina/metabolismo , beta Catenina/genética , Ratas , Proteína Axina/genética , Proteína Axina/metabolismo , Mucosa Bucal/metabolismo , Mucosa Bucal/efectos de los fármacos , Humanos , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/genética , Modelos Animales de Enfermedad
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(5): 930-940, 2024 May 20.
Artículo en Chino | MEDLINE | ID: mdl-38862451

RESUMEN

OBJECTIVE: To explore the pharmacologically active components in areca nut that induce oral submucosal fibrosis (OSF) and the possible mechanism. METHODS: The chemical components in areca nut were analyzed using Thermo QE plus liquid chromatography tandem high-resolution mass spectrometer and Compound discover 3.2 data processing software. The chemical activity of the top 20 compounds was analyzed based on Chinese Pharmacopoeia (2015), PubChem, Chemical book, and SciFinder databases. The potential active components, core targets, biological functions and signaling pathways affecting OSF were analyzed by network pharmacology. The targets of OSF were obtained by integrating Genecards and KEGG databases. The compounds acting on the targets were selected from the Systematic Pharmacology Technology Platform of Traditional Chinese Medicine (TCMSP), and the target-compound, compound-TCM, target-compound-TCM network was constructed. Molecular docking was used to analyze the component-target binding. Immunohistochemistry was used to examine the expressions of key proteins in the PI3K-Akt and MAPK pathways in clinical samples of OSF. RESULTS: The core intersection target genes between the top 10 active ingredients in areca nut extract and OSF involved mainly the PI3K-Akt and MAPK pathways. In the clinical samples, the expressions of PI3K protein decreased and the expressions p-PI3K, AKT1 and PAkt all increased significantly in OSF tissue, where increased JNK protein expression and enhanced activity of c-Jun and c-Fos transcriptional factors were also detected. The OSF patients had significantly elevated plasma levels of IL-6 and IL-8 compared with healthy individuals. CONCLUSION: The main active ingredients including arecoline, arecaine, and guvacine are capable of activating the PI3K-Akt and MAPK pathways to promote the expressions of inflammatory mediators IL-6 and IL-8 and induce collagen hyperplasia, thus leading to the occurrence of oral submucosal fibrosis.


Asunto(s)
Areca , Farmacología en Red , Areca/química , Humanos , Fibrosis de la Submucosa Bucal/metabolismo , Simulación del Acoplamiento Molecular , Transducción de Señal/efectos de los fármacos , Nueces/química , Medicina Tradicional China/métodos , Fosfatidilinositol 3-Quinasas/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Medicamentos Herbarios Chinos/farmacología
9.
Int J Oral Sci ; 16(1): 48, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38897993

RESUMEN

Oral submucous fibrosis (OSF) is a chronic and inflammatory mucosal disease caused by betel quid chewing, which belongs to oral potentially malignant disorders. Abnormal fibroblast differentiation leading to disordered collagen metabolism is the core process underlying OSF development. The epithelium, which is the first line of defense against the external environment, can convert external signals into pathological signals and participate in the remodeling of the fibrotic microenvironment. However, the specific mechanisms by which the epithelium drives fibroblast differentiation remain unclear. In this study, we found that Arecoline-exposed epithelium communicated with the fibrotic microenvironment by secreting exosomes. MiR-17-5p was encapsulated in epithelial cell-derived exosomes and absorbed by fibroblasts, where it promoted cell secretion, contraction, migration and fibrogenic marker (α-SMA and collagen type I) expression. The underlying molecular mechanism involved miR-17-5p targeting Smad7 and suppressing the degradation of TGF-ß receptor 1 (TGFBR1) through the E3 ubiquitination ligase WWP1, thus facilitating downstream TGF-ß pathway signaling. Treatment of fibroblasts with an inhibitor of miR-17-5p reversed the contraction and migration phenotypes induced by epithelial-derived exosomes. Exosomal miR-17-5p was confirmed to function as a key regulator of the phenotypic transformation of fibroblasts. In conclusion, we demonstrated that Arecoline triggers aberrant epithelium-fibroblast crosstalk and identified that epithelial cell-derived miR-17-5p mediates fibroblast differentiation through the classical TGF-ß fibrotic pathway, which provided a new perspective and strategy for the diagnosis and treatment of OSF.


Asunto(s)
Arecolina , Células Epiteliales , Exosomas , Fibroblastos , MicroARNs , Fibrosis de la Submucosa Bucal , Receptor Tipo I de Factor de Crecimiento Transformador beta , MicroARNs/metabolismo , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Humanos , Fibroblastos/metabolismo , Arecolina/farmacología , Células Epiteliales/metabolismo , Exosomas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Proteína smad7/metabolismo , Diferenciación Celular , Transducción de Señal , Movimiento Celular , Ubiquitina-Proteína Ligasas/metabolismo , Areca/efectos adversos
10.
J Oral Pathol Med ; 53(7): 468-479, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38802299

RESUMEN

BACKGROUND: circRNAs have been shown to participate in diverse diseases; however, their role in oral submucous fibrosis (OSF), a potentially malignant disorder, remains obscure. Our preliminary experiments detected the expression of circRNA mitochondrial translation optimization 1 homologue (circMTO1) in OSF tissues (n = 20) and normal mucosa tissues (n = 20) collected from Hunan Xiangya Stomatological Hospital, and a significant decrease of circMTO1 expression was showed in OSF tissues. Therefore, we further explored circMTO1 expression in OSF. METHODS: Target molecule expression was detected using RT-qPCR and western blotting. The migration and invasion of buccal mucosal fibroblasts (BMFs) were assessed using wound healing and Transwell assays. The interaction between miR-30c-5p, circMTO1, and SOCS3 was evaluated using dual luciferase, RNA immunoprecipitation (RIP), and RNA pull-down assays. The colocalisation of circMTO1 and miR-30c-5p was observed using fluorescence in situ hybridisation (FISH). RESULTS: circMTO1 and SOCS3 expression decreased, whereas miR-30c-5p expression increased in patients with OSF and arecoline-stimulated BMFs. Overexpression of circMTO1 effectively restrained the fibroblast-myofibroblast transition (FMT), as evidenced by the increase in expression of Coll I, α-SMA, Vimentin, and the weakened migration and invasion functions in BMFs. Mechanistic studies have shown that circMTO1 suppresses FMT by enhancing SOCS3 expression by sponging miR-30c-5p and subsequently inactivating the FAK/PI3K/AKT pathway. FMT induced by SOCS3 silencing was reversed by the FAK inhibitor TAE226 or the PI3K inhibitor LY294002. CONCLUSION: circMTO1/miR-30c-5p/SOCS3 axis regulates FMT in arecoline-treated BMFs via the FAK/PI3K/AKT pathway. Expanding the sample size and in vivo validation could further elucidate their potential as therapeutic targets for OSF.


Asunto(s)
Fibroblastos , MicroARNs , Fibrosis de la Submucosa Bucal , ARN Circular , Proteína 3 Supresora de la Señalización de Citocinas , Humanos , MicroARNs/metabolismo , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Fibroblastos/metabolismo , ARN Circular/genética , Miofibroblastos , Masculino , Movimiento Celular , Mucosa Bucal/metabolismo , Mucosa Bucal/citología , Mucosa Bucal/patología , Transducción de Señal , Femenino , Células Cultivadas
11.
Stem Cell Res Ther ; 15(1): 113, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38650025

RESUMEN

BACKGROUND: Oral submucous fibrosis (OSF) is a precancerous lesion characterized by fibrous tissue deposition, the incidence of which correlates positively with the frequency of betel nut chewing. Prolonged betel nut chewing can damage the integrity of the oral mucosal epithelium, leading to chronic inflammation and local immunological derangement. However, currently, the underlying cellular events driving fibrogenesis and dysfunction are incompletely understood, such that OSF has few treatment options with limited therapeutic effectiveness. Dental pulp stem cells (DPSCs) have been recognized for their anti-inflammatory and anti-fibrosis capabilities, making them promising candidates to treat a range of immune, inflammatory, and fibrotic diseases. However, the application of DPSCs in OSF is inconclusive. Therefore, this study aimed to explore the pathogenic mechanism of OSF and, based on this, to explore new treatment options. METHODS: A human cell atlas of oral mucosal tissues was compiled using single-cell RNA sequencing to delve into the underlying mechanisms. Epithelial cells were reclustered to observe the heterogeneity of OSF epithelial cells and their communication with immune cells. The results were validated in vitro, in clinicopathological sections, and in animal models. In vivo, the therapeutic effect and mechanism of DPSCs were characterized by histological staining, immunohistochemical staining, scanning electron microscopy, and atomic force microscopy. RESULTS: A unique epithelial cell population, Epi1.2, with proinflammatory and profibrotic functions, was predominantly found in OSF. Epi1.2 cells also induced the fibrotic process in fibroblasts by interacting with T cells through receptor-ligand crosstalk between macrophage migration inhibitory factor (MIF)-CD74 and C-X-C motif chemokine receptor 4 (CXCR4). Furthermore, we developed OSF animal models and simulated the clinical local injection process in the rat buccal mucosa using DPSCs to assess their therapeutic impact and mechanism. In the OSF rat model, DPSCs demonstrated superior therapeutic effects compared with the positive control (glucocorticoids), including reducing collagen deposition and promoting blood vessel regeneration. DPSCs mediated immune homeostasis primarily by regulating the numbers of KRT19 + MIF + epithelial cells and via epithelial-stromal crosstalk. CONCLUSIONS: Given the current ambiguity surrounding the cause of OSF and the limited treatment options available, our study reveals that epithelial cells and their crosstalk with T cells play an important role in the mechanism of OSF and suggests the therapeutic promise of DPSCs.


Asunto(s)
Células Epiteliales , Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/metabolismo , Animales , Células Epiteliales/metabolismo , Linfocitos T/metabolismo , Linfocitos T/inmunología , Ratas , Células Madre/metabolismo , Células Madre/citología , Masculino , Mucosa Bucal/patología , Mucosa Bucal/metabolismo , Comunicación Celular
12.
J Cancer Res Ther ; 20(2): 706-711, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38687943

RESUMEN

BACKGROUND: Oral submucous fibrosis (OSF) is a precancerous lesion, with oral squamous cell carcinoma (OSCC) being the most prevalent malignancy affecting the oral mucosa. The malignant transformation of OSF into OSCC is estimated to occur in 7-13% of cases. Myofibroblasts (MFs) play pivotal roles in both physiological and pathological processes, such as wound healing and tumorigenesis, respectively. This study aimed to explore the involvement of MFs in the progression of OSF and its malignant transformation. MATERIALS AND METHODS: In total, 94 formalin-fixed paraffin-embedded tissue blocks were collected, including normal oral mucosa (NOM; n = 10), early-moderate OSF (EMOSF; n = 29), advanced OSF (AOSF; n = 29), paracancerous OSF (POSF; n = 21), and OSCC (n = 5) samples. Alpha-smooth muscle actin was used for the immunohistochemical identification of MFs. RESULTS: NOM exhibited infrequent expression of MFs. A higher staining index of MFs was found in AOSF, followed by EMOSF and NOM. Additionally, a significant increase in the staining index of MFs was found from EMOSF to POSF and OSCC. The staining index of MFs in NOM, EMOSF, AOSF, POSF, and OSCC was 0.14 ± 0.2, 1.69 ± 1.4, 2.47 ± 1.2, 3.57 ± 2.6, and 8.86 ± 1.4, respectively. All results were statistically significant (P < 0.05). CONCLUSIONS: The expression of MFs exhibited a gradual increase as the disease progressed from mild to malignant transformation, indicating the contributory role of MFs in the fibrogenesis and potential tumorigenesis associated with OSF.


Asunto(s)
Transformación Celular Neoplásica , Inmunohistoquímica , Neoplasias de la Boca , Miofibroblastos , Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/metabolismo , Miofibroblastos/patología , Miofibroblastos/metabolismo , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/metabolismo , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Masculino , Femenino , Mucosa Bucal/patología , Mucosa Bucal/metabolismo , Lesiones Precancerosas/patología , Lesiones Precancerosas/metabolismo , Persona de Mediana Edad , Adulto , Actinas/metabolismo , Progresión de la Enfermedad
13.
Arch Oral Biol ; 162: 105944, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38492547

RESUMEN

OBJECTIVE: Oral submucous fibrosis is a frequently reported potentially malignant disorder characterized by fibrosis and a malignant transformation rate of 7-30%. The role of hypoxia-inducible factor-1/2α in malignant transformation mechanisms of oral submucous fibrosis remains uncharted territory owing to a scarcity of studies. Thus the present systematic review and meta-analysis aimed to determine the role of hypoxia-inducible factor-1/2α in the progression of fibrosis of oral submucous fibrosis and its malignant transformation. MATERIAL AND METHODS: Using PubMed, Google Scholar, and Cochrane Library databases, full-text articles that investigated hypoxia-inducible factor-1/2α in oral submucous fibrosis were entailed for review. A modified Newcastle-Ottawa scale was employed to evaluate risk of bias in all articles and Review Manager was utilized for meta-analysis. RESULTS: Eighteen and eight qualified articles respectively were included for qualitative and quantitative data synthesis. Progressive upregulation of hypoxia-inducible factor-1/2α in oral submucous fibrosis is associated with fibrosis-induced carcinogenesis. A Random-effects model uncloaked that oral submucous fibrosis cases with significantly increased expression of hypoxia-inducible factor-1α had an increased associated risk of malignant transformation compared with controls (combined odds ratio 523.83, 95% confidence interval 125.74- 2182.28, p < 0.00001). CONCLUSION: The existing evidence substantiates the notion that hypoxia-inducible factor-1/2α, a fundamental pathogenetic mechanism of progression and malignant transformation of oral submucous fibrosis in the background of fibrosis.


Asunto(s)
Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia , Transformación Celular Neoplásica , Fibrosis , Hipoxia
14.
Tissue Cell ; 87: 102337, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38430849

RESUMEN

OBJECTIVE: This study aimed to investigate the change and pathological significance of glycogen content in oral squamous cell carcinoma (OSCC) and oral submucous fibrosis (OSF). METHODS AND MATERIALS: 13 normal oral mucosa (NOM), 12 OSF mucosa, and 35 pairs of OSCC tissues and their corresponding adjacent mucosa tissues (AT) were collected from Xiangya Hospital for PAS staining to detect glycogen. Transcriptome sequencing data from OSCC were used to compare glycogen metabolism gene expression differences. Kaplan-Meier method was conducted to estimate Recurrence-free survival (RFS). RESULTS: Glycogen levels were lower in OSF than in NOM and lower in OSCC than in AT. Transcriptome sequencing data analysis showed the expression of most glycogenolysis genes was increased and the expression of glycogen synthesis genes including PPP1R3C and GBE1 was decreased in OSCC tissues. High glycogen level was correlated with poor prognosis in OSCC patients under the background of OSF. CONCLUSION: Glycogen may be used as a potential diagnostic biomolecule for OSF and OSCC, as well as a potential prognostic factor for OSCC in the context of OSF.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello , Neoplasias de la Boca/patología
15.
Int J Biol Macromol ; 264(Pt 1): 130504, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38442830

RESUMEN

Long non-coding RNA FENDRR possesses both anti-fibrotic and anti-cancer properties, but its significance in the development of premalignant oral submucous fibrosis (OSF) remains unclear. Here, we showed that FENDRR was downregulated in OSF specimens and fibrotic buccal mucosal fibroblasts (fBMFs), and overexpression of FENDRR mitigated various myofibroblasts hallmarks, and vice versa. In the course of investigating the mechanism underlying the implication of FENDRR in myofibroblast transdifferentiation, we found that FENDRR can directly bind to miR-214 and exhibit its suppressive effect on myofibroblast activation via titrating miR-214. Moreover, we showed that mitofusin 2 (MFN2), a protein that is crucial to the fusion of mitochondria, was a direct target of miR-214. Our data suggested that FENDRR was positively correlated with MFN2 and MFN2 was required for the inhibitory property of FENDRR pertaining to myofibroblast phenotypes. Additionally, our results showed that the FENDRR/miR-214 axis participated in the arecoline-induced reactive oxygen species (ROS) accumulation and myofibroblast transdifferentiation. Building on these results, we concluded that the aberrant downregulation of FENDRR in OSF may be associated with chronic exposure to arecoline, leading to upregulation of ROS and myofibroblast activation via the miR-214-mediated suppression of MFN2.


Asunto(s)
MicroARNs , Fibrosis de la Submucosa Bucal , Humanos , Miofibroblastos/metabolismo , Arecolina/efectos adversos , Arecolina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Mucosa Bucal/metabolismo , Fibroblastos , MicroARNs/genética , MicroARNs/metabolismo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/farmacología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo
16.
J Oral Biosci ; 66(2): 320-328, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38395254

RESUMEN

BACKGROUND: Oral submucous fibrosis (OSF) is a pathological condition characterized by excessive tissue healing resulting from physical, chemical, or mechanical trauma. Notably, areca nut consumption significantly contributes to the development of oral fibrosis. The current definition of OSF, recognizing its potential for malignant transformation, necessitates a more comprehensive understanding of its pathophysiology and etiology. HIGHLIGHTS: Areca nut induces fibrotic pathways by upregulating inflammatory cytokines such as TGF-ß and expressing additional cytokines. Moreover, it triggers the conversion of fibroblasts to myofibroblasts, characterized by α-SMA and γSMA expression, resulting in accelerated collagen production. Arecoline, a component of areca nut, has been shown to elevate levels of reactive oxygen species, upregulate the expression of various cytokines, and activate specific signaling pathways (MEK, COX2, PI3K), all contributing to fibrosis. Therefore, we propose redefining OSF as "Areca nut-induced oral fibrosis" (AIOF) to align with current epistemology, emphasizing its distinctive association with areca nut consumption. The refined definition enhances our ability to develop targeted interventions, thus contributing to more effective prevention and treatment strategies for oral submucous fibrosis worldwide. CONCLUSION: Arecoline plays a crucial role as a mediator in fibrosis development, contributing to extracellular matrix accumulation in OSF. The re-evaluation of OSF as AIOF offers a more accurate representation of the condition. This nuanced perspective is essential for distinguishing AIOF from other forms of oral fibrosis and advancing our understanding of the disease's pathophysiology.


Asunto(s)
Areca , Arecolina , Fibrosis de la Submucosa Bucal , Fibrosis de la Submucosa Bucal/patología , Fibrosis de la Submucosa Bucal/etiología , Fibrosis de la Submucosa Bucal/metabolismo , Humanos , Areca/efectos adversos , Arecolina/efectos adversos , Citocinas/metabolismo , Transducción de Señal , Nueces/efectos adversos
17.
Eur J Pharmacol ; 967: 176353, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38325798

RESUMEN

Oral submucous fibrosis (OSF) is a chronic oral mucosal disease. The pathological changes of OSF include epithelial damage and subepithelial matrix fibrosis. This study aimed to reveal the epithelial injury mechanism of OSF. A histopathological method was used to analyze oral mucosal tissue from OSF patients and OSF rats. The expression of PDE12 in the oral epithelium was analyzed by immunohistochemistry. The epithelial-mesenchymal transition (EMT) and tight junction proteins in arecoline-treated HOKs were explored by western blotting. Epithelial leakage was assessed by transepithelial electrical resistance and lucifer yellow permeability. The expression of PDE12 and the mitochondrial morphology, mitochondrial permeability transition pore opening, mitochondrial membrane potential, and mitochondrial reactive oxygen species (mtROS) were evaluated in arecoline-induced HOKs. Oxidative phosphorylation (OXPHOS) complexes and ATP content were also explored in HOKs. The results showed significant overexpression of PDE12 in oral mucosal tissue from OSF patients and rats. PDE12 was also overexpressed and aggregated in mitochondria in arecoline-induced HOKs, resulting in dysfunction of OXPHOS and impaired mitochondrial function. An EMT, disruption of tight junctions with epithelial leakage, and extracellular matrix remodeling were also observed. PDE12 overexpression induced by PDE12 plasmid transfection enhanced the mtROS level and interfered with occludin protein localization in HOKs. Interestingly, knockdown of PDE12 clearly ameliorated arecoline-induced mitochondrial dysfunction and epithelial barrier dysfunction in HOKs. Therefore, we concluded that overexpression of PDE12 impaired mitochondrial OXPHOS and mitochondrial function and subsequently impaired epithelial barrier function, ultimately leading to OSF. We suggest that PDE12 may be a new potential target against OSF.


Asunto(s)
Enfermedades Mitocondriales , Fibrosis de la Submucosa Bucal , Animales , Humanos , Ratas , Arecolina/efectos adversos , Arecolina/metabolismo , Mitocondrias , Enfermedades Mitocondriales/metabolismo , Fibrosis de la Submucosa Bucal/inducido químicamente , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Fosforilación Oxidativa
18.
Mol Biol Rep ; 51(1): 303, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38356030

RESUMEN

BACKGROUND: This study aimed to assess silymarin's anticancer and antifibrotic potential through in silico analysis and investigate its impact on in vitro arecoline-induced fibrosis in primary human buccal fibroblasts (HBF). METHODS & RESULTS: The study utilized iGEMDOCK for molecular docking, evaluating nine bioflavonoids, and identified silymarin and baicalein as the top two compounds with the highest target affinity, followed by subsequent validation through a 100ns Molecular Dynamic Simulation demonstrating silymarin's stable behavior with Transforming Growth Factor Beta. HBF cell lines were developed from tissue samples obtained from patients undergoing third molar extraction. Arecoline, a known etiological factor in oral submucous fibrosis (OSMF), was employed to induce fibrogenesis in these HBFs. The inhibitory concentration (IC50) of arecoline was determined using the MTT assay, revealing dose-dependent cytotoxicity of HBFs to arecoline, with notable cytotoxicity observed at concentrations exceeding 50µM. Subsequently, the cytotoxicity of silymarin was assessed at 24 and 72 h, spanning concentrations from 5µM to 200µM, and an IC50 value of 143µM was determined. Real-time polymerase chain reaction (qPCR) was used to analyze the significant downregulation of key markers including collagen, epithelial-mesenchymal transition (EMT), stem cell, hypoxia, angiogenesis and stress markers in silymarin-treated arecoline-induced primary buccal fibroblast cells. CONCLUSION: Silymarin effectively inhibited fibroblast proliferation and downregulated genes associated with cancer progression and EMT pathway, both of which are implicated in malignant transformation. To our knowledge, this study represents the first exploration of silymarin's potential as a novel therapeutic agent in an in vitro model of OSMF.


Asunto(s)
Arecolina , Fibrosis de la Submucosa Bucal , Humanos , Arecolina/efectos adversos , Arecolina/metabolismo , Mucosa Bucal/metabolismo , Simulación del Acoplamiento Molecular , Fibrosis de la Submucosa Bucal/inducido químicamente , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Fibrosis de la Submucosa Bucal/metabolismo , Fibroblastos/metabolismo , Fibrosis
19.
Odontology ; 112(3): 811-825, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38217790

RESUMEN

Oral submucous fibrosis (OSF) is an oral condition characterized by chronic progression, which may lead to the development of malignancy. Currently, available treatments for OSF only provide temporary relief of symptoms, and there is a limited availability of effective interventions that can effectively cure this condition. In this study, we aimed to investigate whether adiponectin (APN) could ameliorate OSF and the mechanisms involved in it. First, human oral mucosal fibroblasts (HOMFs) were cultured, an OSF model was established using arecoline, and APN and Imiquimod treatment were administered. Then we overexpressed NLRP3 and knocked down FOXO3A. FOXO3A, fibrosis-related factors (ɑ-SMA, COL1A, CTGF), TGF-ß1/Smad3 signaling-related factors (TGF-ß1, p-Smad3, Smad3), NLRP3 inflammasome-related factors (NLRP3, Caspase-1, IL-1ß), and ROS levels were evaluated. Finally, we explored the effect of APN on OSF in mice by in vivo experiments. We found that arecoline significantly increased ɑ-SMA, COL1A, CTGF, and TGF-ß1 expressions and promoted Smad3 phosphorylation, while APN significantly inhibited the elevation of these fibrosis-related factors. ROS production was significantly elevated in HOMFs after arecoline treatment, while APN treatment inhibited ROS production. However, the addition of Imiquimod and overexpression of NLRP3 exhibited a trend of elevated ROS, resisting the inhibitory effect of APN. Furthermore, adding Imiquimod and overexpression of NLRP3 elevated ɑ-SMA, COL1A and CTGF and activated TGF-ß1/Smad3 signaling pathway. Additionally, knockdown of FOXO3A enhanced APN-inhibited ɑ-SMA and COL1A. In vivo experiments further confirmed that APN ameliorated OSF in mice by inhibiting ROS/NLRP3 inflammatory pathway. In conclusion, APN ameliorated arecoline-induced OSF by promoting FOXO3A expression and downregulating the ROS/NLRP3 pathway.


Asunto(s)
Arecolina , Modelos Animales de Enfermedad , Proteína Forkhead Box O3 , Proteína con Dominio Pirina 3 de la Familia NLR , Fibrosis de la Submucosa Bucal , Especies Reactivas de Oxígeno , Transducción de Señal , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteína Forkhead Box O3/metabolismo , Ratones , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Fibrosis de la Submucosa Bucal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Arecolina/farmacología , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Células Cultivadas , Proteína smad3/metabolismo , Colágeno Tipo I/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Imiquimod , Masculino , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Western Blotting
20.
Adv Sci (Weinh) ; 11(12): e2306515, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38229179

RESUMEN

In South and Southeast Asia, the habit of chewing betel nuts is prevalent, which leads to oral submucous fibrosis (OSF). OSF is a well-established precancerous lesion, and a portion of OSF cases eventually progress to oral squamous cell carcinoma (OSCC). However, the specific molecular mechanisms underlying the malignant transformation of OSCC from OSF are poorly understood. In this study, the leading-edge techniques of Spatial Transcriptomics (ST) and Spatial Metabolomics (SM) are integrated to obtain spatial location information of cancer cells, fibroblasts, and immune cells, as well as the transcriptomic and metabolomic landscapes in OSF-derived OSCC tissues. This work reveals for the first time that some OSF-derived OSCC cells undergo partial epithelial-mesenchymal transition (pEMT) within the in situ carcinoma (ISC) region, eventually acquiring fibroblast-like phenotypes and participating in collagen deposition. Complex interactions among epithelial cells, fibroblasts, and immune cells in the tumor microenvironment are demonstrated. Most importantly, significant metabolic reprogramming in OSF-derived OSCC, including abnormal polyamine metabolism, potentially playing a pivotal role in promoting tumorigenesis and immune evasion is discovered. The ST and SM data in this study shed new light on deciphering the mechanisms of OSF-derived OSCC. The work also offers invaluable clues for the prevention and treatment of OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Transcriptoma , Microambiente Tumoral , Transformación Celular Neoplásica , Perfilación de la Expresión Génica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...