Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Tissue Eng Regen Med ; 21(1): 123-135, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37755664

RESUMEN

BACKGROUND: Oral submucous fibrosis (OSF) is a chronic disease with carcinogenic tendency that poses a non-negligible threat to human health. Exosomes derived from human adipose mesenchymal stem cells (ADSC-Exo) reduces visceral and cutaneous fibroses, but their role in OSF has received little attention. The aim of this study was to investigate the effects of ADSC-Exo on OSF and elucidate the mechanism. METHODS: In brief, ADSCs were extracted from adipose tissues and subjected to flow cytometry and induction culture. Fibroblasts were isolated from human buccal mucosa and subjected to immunofluorescence. Myofibroblasts were obtained from fibroblasts induced by arecoline and identified. Immunofluorescence assay confirmed that myofibroblasts could take up ADSC-Exo. The effects of ADSC-Exo on the proliferative and migratory capacities of myofibroblasts were examined using the Cell Counting Kit-8 and scratch assay. Real-time quantitative polymerase chain reaction (qPCR) was performed to evaluate mothers against decapentaplegic homolog 2 (Smad2), Smad3, Smad7, collagen type 1 (Col1), Col3, alpha smooth muscle actin (α-SMA), fibronectin, and vimentin. Western blotting was performed to detect phospho (p)-Smad2, Smad2, p-Smad2/3, Smad2/3, Smad7, Col1, Col3, α-SMA, fibronectin, and vimentin. Furthermore, the dual-luciferase reporter assay was performed to prove that miR-181a-5p in ADSC-Exo directly inhibited the expression of Smad2 mRNA to regulate the transforming growth factor beta (TGF-ß) pathway. We also performed qPCR and western blotting to verify the results. RESULTS: ADSC-Exo could promote the proliferation and migration of myofibroblasts, reduce the expressions of p-smad2, Smad2, p-smad2/3, Smad2/3, Col1, αSMA, fibronectin, and vimentin and elevated the levels of Smad7 and Col3. In addition, miR-181a-5p was highly expressed in ADSC-Exo and bound to the 3'-untranslated region of Smad2. ADSC-Exo enriched with miR-181a-5p reduced collagen production in myofibroblasts and modulated the TGF-ß pathway. CONCLUSIONS: ADSC-Exo promoted the proliferative and migratory capacities of myofibroblasts and inhibited collagen deposition and trans-differentiation of myofibroblasts in vitro. miR-181a-5p in exosomes targets Smad2 to regulate the TGF-ß pathway in myofibroblasts. ADSC-Exo perform antifibrotic actions through the miR-181a-5p/Smad2 axis and may be a promising clinical treatment for OSF.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , MicroARNs , Fibrosis de la Submucosa Bucal , Humanos , Colágeno Tipo I/metabolismo , Exosomas/metabolismo , Fibronectinas/metabolismo , Fibrosis , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/terapia , Fibrosis de la Submucosa Bucal/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vimentina/metabolismo
10.
Drug Des Devel Ther ; 17: 3085-3101, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37854130

RESUMEN

Purpose: Arecoline is one of the main toxic components of arecoline to cause oral mucosal lesions or canceration, which seriously affects the survival and life quality of patients. This study analyzed the mechanism of Jiawei Danxuan Koukang (JDK) in alleviating arecoline induced oral mucosal lesions, to provide new insights for the treatment of oral submucosal fibrosis (OSF) or cancerosis. Methods: Metabolomics was applied to analyze the composition of JDK and serum metabolites. The active ingredients of JDK were analyzed by the combined ultra-high performance liquid chromatography and mass spectrometry. The target network of JDK, metabolites and OSF was analyzed by network pharmacology, and molecular docking. Oral mucosal lesions and fibrosis were analyzed by HE and Masson staining. Cell differentiation, proliferation and apoptosis were detected. The expressions of α-SMA, Collagen I, Vimentin, Snail, E-cadherin, AR and NOTCH1 were detected by Western blot. Results: Arecoline induced the gradual atrophy and thinning of rat oral mucosal, collagen accumulation, the increase expressions of fibrosis-related proteins and Th17/Treg ratio. JDK inhibited arecoline-induced oral mucosal lesions and inflammatory infiltration. Arecoline induced changes of serum metabolites in Aminoacyl-tRNA biosynthesis, Alanine, aspartate and glutamate metabolism and Arginine biosynthesis pathways, which were reversed by M-JDK. Quercetin and AR were the active ingredients and key targets of JDK, metabolites and OSF interaction. Arecoline promoted the expression of AR protein, and the proliferation of oral fibroblasts. Quercetin inhibited the effect of arecoline on oral fibroblasts, but was reversed by AR overexpression. Arecoline induced NOTCH1 expression in CAL27 and SCC-25 cells, and promoted cell proliferation, but was reversed by M-JDK or quercetin. Conclusion: JDK improved the arecoline-induced OSF and serum metabolite functional pathway. Quercetin targeted AR protein to improve arecoline-induced OSF. JDK and quercetin inhibited arecoline-induced NOTCH1 protein expression in CAL27 and SCC-25 cells to play an anti-oral cancer role.


Asunto(s)
Arecolina , Fibrosis de la Submucosa Bucal , Humanos , Ratas , Animales , Arecolina/efectos adversos , Cromatografía Líquida de Alta Presión , Farmacología en Red , Simulación del Acoplamiento Molecular , Quercetina/farmacología , Fibrosis de la Submucosa Bucal/etiología , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Mucosa Bucal/patología , Fibroblastos , Colágeno/farmacología , Fibrosis , Espectrometría de Masas
11.
Am J Pathol ; 193(9): 1208-1222, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37328100

RESUMEN

Oral submucous fibrosis (OSF) is a potentially malignant disorder of the oral mucosa; however, whether and how the fibrotic matrix of OSF is involved in the malignant transformation of epithelial cells remains unknown. Herein, oral mucosa tissue from patients with OSF, OSF rat models, and their controls were used to observe the extracellular matrix changes and epithelial-mesenchymal transformation (EMT) in fibrotic lesions. Compared with controls, oral mucous tissues from patients with OSF showed an increased number of myofibroblasts, a decreased number of blood vessels, and increased type I and type III collagen levels. In addition, the oral mucous tissues from humans and OSF rats showed increased stiffness, accompanied by increased EMT activities of epithelial cells. The EMT activities of stiff construct-cultured epithelial cells were increased significantly by exogenous piezo-type mechanosensitive ion channel component 1 (Piezo1) activation, and decreased by yes-associated protein (YAP) inhibition. During ex vivo implantation, oral mucosal epithelial cells of the stiff group showed increased EMT activities and increased levels of Piezo1 and YAP compared with those in the sham and soft groups. These results indicate that increased stiffness of the fibrotic matrix in OSF led to increased proliferation and EMT of mucosal epithelial cells, in which the Piezo1-YAP signal transduction is important.


Asunto(s)
Fibrosis de la Submucosa Bucal , Humanos , Ratas , Animales , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Transición Epitelial-Mesenquimal , Miofibroblastos/metabolismo , Células Epiteliales/metabolismo
12.
Inflammopharmacology ; 31(5): 2349-2368, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37106237

RESUMEN

Oral submucous fibrosis (OSF) is a chronic, progressive, and precancerous condition mainly caused by chewing areca nut. Currently, OSF therapy includes intralesional injection of corticosteroids with limited therapeutic success in disease management. Therefore, a combined approach of in silico, in vitro and in vivo drug development can be helpful. Polyphenols are relatively safer than other synthetic counterparts. We used selected polyphenols to shortlist the most suitable compound by in silico tools. Based on the in silico results, epigallocatechin-3-gallate (EGCG), quercetin (QUR), resveratrol, and curcumin had higher affinity and stability with the selected protein targets, transforming growth factor beta-1 (TGF-ß1), and lysyl oxidase (LOX). The efficacy of selected polyphenols was studied in primary buccal mucosal fibroblasts followed by in vivo areca nut extract induced rat OSF model. In in vitro studies, the induced fibroblast cells were treated with EGCG and QUR. EGCG was safer at higher concentrations and more efficient in reducing TGF-ß1, collagen type-1A2 and type-3A1 mRNA expression than QUR. In vivo studies confirmed that the EGCG hydrogel was efficient in improving the disease conditions compared to the standard treatment betamethasone injection with significant reduction in TGF-ß1 and collagen concentrations with increase in mouth opening. EGCG can be considered as a potential, safer and efficient phytomolecule for OSF therapy and its mucoadhesive topical formulation help in the improvement of patient compliance without any side effects. Highlights Potential polyphenols were shortlisted to treat oral submucous fibrosis (OSF) using in silico tools Epigallocatechin 3-gallate (EGCG) significantly reduced TGF-ß1 and collagen both in vitro and in vivo EGCG hydrogel enhanced antioxidant defense, modulated inflammation by reducing TGF-ß1 and improved mouth opening in OSF rat model.


Asunto(s)
Fibrosis de la Submucosa Bucal , Humanos , Animales , Ratas , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Fibrosis de la Submucosa Bucal/inducido químicamente , Fibrosis de la Submucosa Bucal/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Polifenoles/farmacología , Colágeno , Hidrogeles/efectos adversos
13.
PeerJ ; 11: e15158, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37096061

RESUMEN

Objectives: Cellular senescence is strongly associated with fibrosis and tumorigenesis. However, whether the epithelium of oral submucous fibrosis (OSF) undergoes premature senescence remains unclear. This study investigates the roles of senescent epithelial cells in OSF. Methods: The immunohistochemistry and Sudan black B staining were performed to identify epithelium senescence in OSF tissues. Arecoline was used to induce human oral keratinocytes (HOKs) senescence. The cell morphology, senescence-associated ß galactosidase activity, cell counting Kit 8, immunofluorescence, quantitative real-time PCR, and western blot assay were used to identification of senescent HOKs. The enzyme-linked immunosorbent assay was exerted to evaluate the levels of transforming growth factor ß1 (TGF-ß1) in the supernatants of HOKs treated with or without arecoline. Results: The senescence-associated markers, p16 and p21, were overexpressed in OSF epithelium. These expressions were correlated with alpha-smooth actin (α-SMA) positively and proliferating cell nuclear antigen (PCNA) negatively. Moreover, Sudan black staining showed that there was more lipofuscin in OSF epithelium. In vitro, HOKs treated with arecoline showed senescence-associated characteristics including enlarged and flattened morphology, senescence-associated ß galactosidase staining, cell growth arrest, γH2A.X foci, upregulation of p53, p21, and TGF-ß1 protein levels. Moreover, senescent HOKs secreted more TGF-ß1. Conclusions: Senescent epithelial cells are involved in OSF progression and may become a promising target for OSF treatment.


Asunto(s)
Fibrosis de la Submucosa Bucal , Factor de Crecimiento Transformador beta1 , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Fibrosis de la Submucosa Bucal/metabolismo , Arecolina/metabolismo , Queratinocitos/metabolismo , beta-Galactosidasa/metabolismo
14.
Nutrients ; 15(6)2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36986051

RESUMEN

Oral submucous fibrosis (OSF) is a premalignant disorder and persistent activation of myofibroblasts is implicated in this pathological progression. Increasing attention has been addressed towards non-coding RNA-regulated myofibroblasts activities and the effects of phytochemicals on non-coding RNA modulation are of great importance. In the present study, we examined the anti-fibrosis property of α-mangostin, a xanthone isolated from the pericarp of mangosteen. We found that α-mangostin exhibited inhibitory potency in myofibroblast activities and expression of fibrosis markers at the concentrations that caused neglectable damage to normal cells. Apart from the downregulation of TGF-ß1/Smad2 signaling, we found that α-mangostin attenuated the expression of long non-coding RNA LincROR as well. Our results demonstrated that the effects of α-mangostin on myofibroblast activation were reverted when LincROR was overexpressed. Additionally, we showed the expression of LincROR in OSF specimens was elevated and silencing of LincROR successfully attenuated myofibroblast characteristics and TGF-ß1/Smad2 activation. Taken together, these findings indicated that the anti-fibrosis effects of α-mangostin merit consideration and may be due to the attenuation of LincROR.


Asunto(s)
Fibrosis de la Submucosa Bucal , Xantonas , Humanos , Miofibroblastos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Abajo , Xantonas/farmacología , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología
15.
Arch Oral Biol ; 148: 105644, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36804642

RESUMEN

OBJECTIVE: The review aims to comprehend various factors engaged in the alteration of molecular events resulting in Oral submucous fibrosis (OSMF) and its malignant transformation. DESIGN: Literature pertinent to pathways involved in OSMF were explored in databases such as PubMed, Scopus and Google Scholar. The relevant literature was reviewed and critically appraised in this narrative review. RESULTS: Areca nut components influence myriad of cellular molecules such as cytokines, growth factors, myofibroblasts, non-coding RNAs and alter their expression. These aberrantly expressed molecules drive the progression of OSMF from localized inflammation to fibrosis of buccal mucosa. The oral tissue suffers from oxidative stress, hypoxia, autophagy, aberration of cell cycle and DNA damage. Apoptosis of epithelial layer results in its atrophy facilitating deeper penetration of areca nut elements. With the advance of disease, epithelial-mesenchymal transition eventuates and promotes dysplasia. The jeopardized expression of various cellular molecules, suppressed apoptosis, along with increased genetic alterations and neovascularization favors the malignant transformation. CONCLUSION: OSMF is a progressive disorder with complex mechanism of pathogenesis initiated by inflammation of oral mucosa. Continuous habit of areca nut chewing and the resulting insult to the tissues prevents healing process and is destined to debilitating disease which affects the quality of life with a higher probability of progression to malignancy.


Asunto(s)
Neoplasias de la Boca , Fibrosis de la Submucosa Bucal , Fibrosis de la Submucosa Bucal/metabolismo , Calidad de Vida , Mucosa Bucal/metabolismo , Neoplasias de la Boca/patología , Inflamación/patología , Areca
16.
Int J Biol Macromol ; 232: 123400, 2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-36702230

RESUMEN

Long non-coding RNA XIST promotes the development of various types of head and neck cancers, but its role in the progression of precancerous oral submucous fibrosis (OSF) has not been determined yet. As such, we aimed to examine whether XIST implicates in the regulation of myofibroblast activation. Our results showed that the expression of XIST was upregulated in OSF tissues and fibrotic buccal mucosal fibroblasts (fBMFs), and the silencing of XIST downregulated several myofibroblasts features. We demonstrated that elevation of let-7i after inhibition of XIST may lead to reduced myofibroblast activation. On the contrary, overexpression of high mobility group AT-Hook 1 (HMGA1) following the suppression of let-7i may result in enhanced myofibroblast activities. Moreover, we showed that the suppressive effect of silencing of XIST on myofibroblasts hallmarks was reversed by let-7i inhibition or HMGA1 overexpression, suggesting the pro-fibrotic property of XIST was mediated by downregulation of let-7i and upregulation of HMGA1. These findings revealed that myofibroblast activation of fBMFs may attribute to the alteration of the XIST/let-7i/HMGA1 axis. Therapeutic approaches to target this axis may serve as a promising direction to ameliorate the malignant progression of OSF.


Asunto(s)
MicroARNs , Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Miofibroblastos/metabolismo , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Proteína HMGA1a/uso terapéutico , Movimiento Celular , Mucosa Bucal/metabolismo , Factores de Transcripción/metabolismo , MicroARNs/genética
17.
Acta Biomater ; 157: 639-654, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36509401

RESUMEN

Oral submucous fibrosis (OSF) is a chronic, inflammatory and potentially malignant oral disorder. Its pathophysiology is extremely complex, including excessive collagen deposition, massive inflammatory infiltration, and capillary atrophy. However, the existing clinical treatment methods do not fully take into account all the pathophysiological processes of OSF, so they are generally low effective and have many side effects. In the present study, we developed an injectable sodium hyaluronate/45S5 bioglass composite hydrogel (BG/HA), which significantly relieved mucosal pallor and restricted mouth opening in OSF rats without any obvious side effects. The core mechanism of BG/HA in the treatment of OSF is the release of biologically active silicate ions, which inhibit collagen deposition and inflammation, and promote angiogenesis and epithelial regeneration. Most interestingly, silicate ions can overall regulate the physiological environment of OSF by down-regulating α-smooth muscle actin (α-SMA) and CD68 and up-regulating CD31 expression, as well as regulating the expression of pro-fibrotic factors [transforming growth factor-ß1 (TGF-ß1), interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α) and tissue inhibitors of metalloproteinase-1 (TIMP-1)] and anti-fibrotic factors [interleukin-1ß (IL-1ß)] in macrophage. In conclusion, our study shows that BG/HA has great potential in the clinical treatment of OSF, which provides an important theoretical basis for the subsequent development of new anti-fibrotic clinical preparations. STATEMENT OF SIGNIFICANCE: : Oral submucous fibrosis (OSF) is a chronic, inflammatory and potentially malignant mucosal disease with significant impact on the quality of patients' life. However, the existing clinical treatments have limited efficacy and many side effects. There is an urgent need for development of specific drugs for OSF treatment. In the present study, bioglass (BG) composited with sodium hyaluronate solution (HA) was used to treat OSF in an arecoline-induced rat model. BG/HA can significantly inhibit collagen deposition, regulate inflammatory response, promote angiogenesis and repair damaged mucosal epithelial cells, and thereby mitigate the development of fibrosis in vivo.


Asunto(s)
Fibrosis de la Submucosa Bucal , Ratas , Animales , Fibrosis de la Submucosa Bucal/tratamiento farmacológico , Fibrosis de la Submucosa Bucal/inducido químicamente , Fibrosis de la Submucosa Bucal/metabolismo , Mucosa Bucal , Ácido Hialurónico/farmacología , Ácido Hialurónico/metabolismo , Hidrogeles/metabolismo , Colágeno/farmacología , Colágeno/metabolismo
18.
Oral Dis ; 29(4): 1782-1790, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-35150031

RESUMEN

Oral submucous fibrosis (OSF) caused by areca nut chewing is a prevalent fibrotic disease in Asia-Pacific countries. Arecoline-induced migration of fibroblasts (FBs) plays a vital role in the development of OSF. However, the specific molecular mechanisms involved remain unclear. Many studies have shown that tyrosine sulphation of chemokines can influence cell migration. Herein, we demonstrated that arecoline stimulates tyrosine sulphation of the chemokine receptor 4 (CXCR4) through the tyrosylprotein sulphotransferase-1 (TPST-1) to enhance the migration ability of FBs. Moreover, by RNA-Seq analysis, we found that the most significantly altered pathway was the EGFR pathway after the arecoline stimulation for FBs. After the knockdown of arecoline-induced EGFR expression, the tyrosine sulphation of CXCR4 was significantly decreased by the inhibition of TPST-1 induction. Finally, in human OSF specimens, TPST-1 expression was directly correlated with the expression of CXCR4. These data indicate that the arecoline-induced tyrosine sulphation of CXCR4, which is regulated by TPST-1, might be a potential mechanism that contributes to FB migration in OSF.


Asunto(s)
Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/metabolismo , Arecolina/farmacología , Tirosina/efectos adversos , Tirosina/metabolismo , Fibroblastos , Receptores ErbB/metabolismo , Mucosa Bucal/metabolismo , Areca , Receptores CXCR4/metabolismo
19.
Dis Mon ; 69(1): 101351, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35341590

RESUMEN

BACKGROUND: Oral submucous fibrosis (OSMF) is a chronic disease with significantly increasing malignant transformation rate. To date the pathogenesis of OSMF has been considered to be associated with areca nut constituents and their action on fibroblasts. However, fibrosis is also associated with immunological factors such as chemokines. In-depth analysis of such factors is the need of the hour in OSMF to better understand the pathogenesis so that effective therapeutic strategies can be developed in the future. MATERIALS AND METHOD: Clinically diagnosed cases of OSMF (n=21) and healthy individuals (n=10) were enrolled in the present study. Chemokines such as CCL2, CCL3, CCL4, CCL5, CCL11, CCL17, CCL28, CXCL1, CXCL5, CXCL8, CXCL9, CXCL10, and CXCL11 were assessed using the chemokine bead array in conjunction with the flow cytometry, along with real-time PCR (RT-PCR). The transcription factors CREB, NF-κB and NFAT5 were also studied for their expressions. The analysis of pg/ml (picogram/milliliter) values was done by using LEGENDplex™ Data Analysis Software. RESULTS: The results obtained demonstrated early phase transient increase in CXCL-11, CCL20, CXCL9, CCL3, CCL2, CXCL10 and CXCL8. However, the expression of CCL3, CXCL10 and CXCL8 was higher in the late stage as compared to the early stage. The relative gene expression of CREB, NF-κB, NFAT5 were upregulated in the late stage of OSMF when compared to normal. CONCLUSION: Distinctive sets of chemokine expression during the early and late stages of OSMF suggest a unique pattern of disease progression playing an important role in the pathogenesis.


Asunto(s)
Fibrosis de la Submucosa Bucal , Humanos , Fibrosis de la Submucosa Bucal/genética , Fibrosis de la Submucosa Bucal/metabolismo , Factores de Transcripción , FN-kappa B , Progresión de la Enfermedad , Expresión Génica
20.
BMJ Open ; 12(12): e060738, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36523229

RESUMEN

OBJECTIVES: We aim to evaluate salivary matrix metalloproteinases (MMP-8) levels in oral submucous fibrosis (OSF) and oral squamous cell carcinoma (OSCC) for the purpose of diagnosis at the early stage via non-invasive method. SETTING: The study was multicentre, carried out at a tertiary care hospital in Karachi, Pakistan. PARTICIPANTS: A total 60 participants of any age, sex and ethnicity were randomly selected for the purpose of this study. Patients demonstrating clinical evidence of OSF and biopsy-proven cases of OSCC were included. Patients with indeterminate histopathological report, immunodeficiency, autoimmune disorder, chronic medical and periodontal disease (periodontal depth greater than 5 mm) and individuals with interincisal mouth opening greater than 35 mm were excluded from the study. INTERVENTIONS: Salivary MMP-8 levels were observed in OSF, healthy and OSCC groups by using ELISA. One way analysis of variance was applied to establish whether MMP-8 levels of disease-free individuals and patients suffering from OSF and OSCC differed from each other. RESULTS: Statistically significant difference in salivary MMP-8 expression in diseased and control group was observed. MMP-8 levels in OSCC (0.64 ng/mL) and OSF (0.66 ng/mL) were underexpressed as compared with healthy participants (7.9 ng/mL). CONCLUSION: MMP-8 levels were underexpressed in OSCC and OSF patients as compared with controls, which imply that MMP-8 level has an inverse relation with OSCC and OSF.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Fibrosis de la Submucosa Bucal , Humanos , Neoplasias de la Boca/metabolismo , Fibrosis de la Submucosa Bucal/metabolismo , Fibrosis de la Submucosa Bucal/patología , Carcinoma de Células Escamosas/diagnóstico , Metaloproteinasa 8 de la Matriz , Carcinoma de Células Escamosas de Cabeza y Cuello , Estudios Transversales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...