Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cancer Sci ; 113(4): 1220-1234, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35189004

RESUMEN

Owing to recent advances in immunotherapies, the overall survival of patients with skin cutaneous melanoma (SKCM) has increased; however, the 5-year survival rate of metastatic patients remains poor. Skin cutaneous melanoma-upregulated genes were screened via analysis of differentially expressed genes (GSE3189 and GSE46517), and metastasis-related oncogenes were identified via weighted gene coexpression network analysis of the GSE46517 dataset. As confirmed by the Tumor Immune Estimation Resource, we found highly expressed centromere protein F (CENPF) in SKCM and its metastases. Immunostaining of human melanoma tissues demonstrated high CENPF expression. According to the Kaplan-Meier survival curve log-rank test, receiver-operating characteristic curve, and univariate and multivariate analyses, the Cancer Genome Atlas (TCGA) database suggested CENPF be a typical independent predictor of SKCM. The CIBERSORT algorithm classified the types of the immune cells from GSE46517 and showed higher proportion of CD4+ memory-activated T cells in metastatic melanoma. Single-sample gene set enrichment analysis of TCGA data confirmed the correlation between CENPF and activated CD4+ T cells. Centromere protein F was positively correlated with tumor mutational burden and CD4+ memory T cell markers (interleukin [IL]-23A, CD28, and CD62L), negatively associated with memory T cell maintenance factors (IL-7 and IL-15) by correlation analysis. Moreover, immunofluorescence showed high coexpression of CENPF and IL23A, CD4 in melanoma. Upregulated CENPF might lead to premature depletion of CD4+ memory T cells and immunosuppression. Nomogram indicated CENPF clinical predictive value for 1-, 3-, 5-, and 7-year melanoma overall survival. Therefore, CENPF plays a vital role in the progression and metastasis of melanoma and can be an effective therapeutic target.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Biomarcadores de Tumor/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Proteínas Cromosómicas no Histona , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/patología , Células T de Memoria , Proteínas de Microfilamentos , Pronóstico , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
2.
Lab Invest ; 100(5): 751-761, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31925326

RESUMEN

The skin plays a critical role in maintenance of water homeostasis. Dysfunction of the skin barrier causes not only delayed wound healing and hypertrophic scarring, but it also contributes to the development of various skin diseases. Dermatitis is a chronic inflammatory skin disorder that has several different subtypes. Skin of contact dermatitis and atopic dermatitis (AD) show epidermal barrier dysfunction. Nax is a sodium channel that regulates inflammatory gene expression in response to perturbation of barrier function of the skin. We found that in vivo knockdown of Nax using RNAi reduced hyperkeratosis and keratinocyte hyperproliferation in rabbit ear dermatitic skin. Increased infiltration of inflammatory cells (mast cells, eosinophils, T cells, and macrophages), a characteristic of dermatitis, was reduced by Nax knockdown. Upregulation of PAR-2 and thymic stromal lymphopoietin (TSLP), which induce Th2-mediated allergic responses, was inhibited by Nax knockdown. In addition, expression of COX-2, IL-1ß, IL-8, and S100A9, which are downstream genes of Nax and are involved in dermatitis pathogenesis, were also decreased by Nax knockdown. Our data show that knockdown of Nax relieved dermatitis symptoms in vivo and indicate that Nax is a novel therapeutic target for dermatitis, which currently has limited therapeutic options.


Asunto(s)
Dermatitis Atópica , Piel , Canales de Sodio Activados por Voltaje , Animales , Proliferación Celular/genética , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Dermatitis Atópica/fisiopatología , Regulación hacia Abajo/genética , Eosinófilos/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Queratinocitos/metabolismo , Queratosis/genética , Queratosis/patología , Queratosis/fisiopatología , Mastocitos/metabolismo , Conejos , Piel/citología , Piel/patología , Piel/fisiopatología , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo
3.
BMC Cancer ; 20(1): 927, 2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-32993558

RESUMEN

BACKGROUND: Human skin cutaneous melanoma is the most common and dangerous skin tumour, but its pathogenesis is still unclear. Although some progress has been made in genetic research, no molecular indicators related to the treatment and prognosis of melanoma have been found. In various diseases, dysregulation of lncRNA is common, but its role has not been fully elucidated. In recent years, the birth of the "competitive endogenous RNA" theory has promoted our understanding of lncRNAs. METHODS: To identify the key lncRNAs in melanoma, we reconstructed a global triple network based on the "competitive endogenous RNA" theory. Gene Ontology and KEGG pathway analysis were performed using DAVID (Database for Annotation, Visualization, and Integration Discovery). Our findings were validated through qRT-PCR assays. Moreover, to determine whether the identified hub gene signature is capable of predicting the survival of cutaneous melanoma patients, a multivariate Cox regression model was performed. RESULTS: According to the "competitive endogenous RNA" theory, 898 differentially expressed mRNAs, 53 differentially expressed lncRNAs and 16 differentially expressed miRNAs were selected to reconstruct the competitive endogenous RNA network. MALAT1, LINC00943, and LINC00261 were selected as hub genes and are responsible for the tumorigenesis and prognosis of cutaneous melanoma. CONCLUSIONS: MALAT1, LINC00943, and LINC00261 may be closely related to tumorigenesis in cutaneous melanoma. In addition, MALAT1 and LINC00943 may be independent risk factors for the prognosis of patients with this condition and might become predictive molecules for the long-term treatment of melanoma and potential therapeutic targets.


Asunto(s)
Carcinogénesis/genética , Melanoma/genética , ARN Largo no Codificante/genética , Neoplasias Cutáneas/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Estimación de Kaplan-Meier , Masculino , MicroARNs/genética , Pronóstico , ARN Largo no Codificante/clasificación , ARN Mensajero/genética , Melanoma Cutáneo Maligno
4.
Exp Dermatol ; 28(5): 576-584, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30903711

RESUMEN

Nax is an atypical sodium channel that mediates inflammatory pathways in pathological conditions of the skin. In this study, we developed a skin inflammation model in the rabbit ear through application of imiquimod (IMQ). Knockdown of Nax using RNAi attenuated IMQ-induced skin inflammation, including skin erythema, scaling and papule formation. Histologic analysis showed that thickening and insufficient differentiation of the epidermis found in psoriasis-like skin were normalized by administration of Nax -RNAi. Excessive infiltration of inflammatory cells found in inflammatory lesions, such as mast cells, eosinophils, neutrophils, T cells and macrophages, was reduced by Nax -RNAi. Expression of S100A9, which is a downstream gene of Nax and a mediator of inflammation, was decreased by Nax -RNAi. Our results demonstrated that knockdown of Nax ameliorated IMQ-induced psoriasis-like skin inflammation in vivo. Thus, targeting of Nax may represent a potential therapeutic option for the treatment of psoriasis.


Asunto(s)
Dermatitis/tratamiento farmacológico , Imiquimod/farmacología , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Canales de Sodio Activados por Voltaje/genética , Animales , Epidermis/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Inflamación/inducido químicamente , Psoriasis/inducido químicamente , Interferencia de ARN , Conejos , Piel/patología
5.
BMC Cancer ; 19(1): 609, 2019 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-31226958

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is the major subtype of esophageal cancer with high aggressiveness and poor prognosis. There is an urgent need for understanding the molecular mechanism underlying the development and progression of ESCC. METHODS: ESCC tissues and corresponding non-neoplastic tissues were collected. The expression and function of miR-124-3p and BCAT1 in two cell lines KYSE-150 and Eca109 were determined. RESULTS: We show downregulation of miR-124-3p expression in ESCC tissues, which is highly correlated with proliferation and migration of ESCC cell lines KYSE-150 and Eca109. miR-124-3p show high correlation with TNM stage and differentiation grade. Furthermore, miR-124-3p directly targets mRNA 3'UTR region of BCAT1, which results in upregulation of BCAT1 expression as observed in ESCC tissues and cell lines. Also, our data indicates that BCAT1 high expression is strongly linked to the disease-free survival, tumor size, pathologic stage, T classification and differentiation grade. On the other hand, we clarified the upstream mechanism regulating miR-124-3p expression in ESCC, which involves in the hypermethylation-silencing regulation mediated by DNA methyltransferase 1(DNMT1), which is of high expression in ESCC tissues and cell lines in the present study. In addition, DNMT1 knockdown or inhibition of DNMT1 function contributes to downregulation of miR-124-3p and BCAT1 expression. CONCLUSIONS: Our study thus clarifies a new mechanism that DNMT1/miR-124/BCAT1 axis regulates the development and progression of ESCC.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Transaminasas/metabolismo , Regiones no Traducidas 3' , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica/fisiopatología , Estadificación de Neoplasias , Transaminasas/genética , Transfección
6.
Cell Physiol Biochem ; 49(6): 2333-2347, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30261495

RESUMEN

BACKGROUND/AIMS: Little is known how miR-203 is involved in epidermal stem cells (ESCs) differentiation and scar formation. METHODS: We first used luciferase assay to determine the interaction of miR-203 with the 3'-UTR in regulation of Hes1 expression. We then used flow cytometry to analyze the effects of miR-203 expression on the differentiation of ESCs to MFB by determination of CK15 ratio and α-SMA. To confirm the results of flow cytometry analysis, we used Western blot to examine the expression of α-SMA, Collagen I (Col I), and Collagen III (Col III), as well as the expression of Notch1, Jagged1, and Hes1 in ESCs after the treatment of pre-miR-203 or anti-miR-203. Finally, we examined the effects local anti-miR-203 treatment on would closure and scar formation using a mouse skin wound model. RESULTS: Pre-miR-203 treatment increased ESCs differentiation to MFB cells, as indicated by decreased CK15 ratio and increased MFB biomarkers. This phenomenon was reversed by overexpression of Hes1 in ESCs. In addition, skin incision increased expression of miR-203 in wound tissue. Local treatment of anti-miR-203 could accelerate wound closure and reduce scar formation in vivo, which was associated with increased re-epithelialization, skin attachment regeneration, and collagen reassignment. Finally, we confirmed that anti-miR-203 treatment could inhibit ESCs differentiation in vivo via increasing Hesl expression. CONCLUSION: Taken together, our results suggested that overexpression of miR-203 in ESCs after skin wound may be a critical mechanism underlying the scar formation.


Asunto(s)
Cicatriz/prevención & control , MicroARNs/metabolismo , Factor de Transcripción HES-1/metabolismo , Cicatrización de Heridas , Regiones no Traducidas 3' , Actinas/metabolismo , Animales , Antagomirs/metabolismo , Diferenciación Celular , Cicatriz/patología , Células Epidérmicas , Femenino , Hiperplasia/patología , Queratina-15/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Miofibroblastos/citología , Miofibroblastos/metabolismo , Piel/patología , Células Madre/citología , Células Madre/metabolismo , Factor de Transcripción HES-1/antagonistas & inhibidores , Factor de Transcripción HES-1/genética
7.
Am J Pathol ; 186(1): 109-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26597884

RESUMEN

The most critical function of the epidermis is to prevent water loss and maintain skin homeostasis. Disruption of the functional skin barrier causes delayed wound healing, hypertrophic scarring, and many skin diseases. Herein, we show that reduced hydration increases the expression of S100 protein family members, S100A8/S100A9, in stratified keratinocyte culture and human ex vivo skin culture. Immunohistological analyses show that S100A8/A9 are highly expressed in the epidermis of human hypertrophic scar and keloid tissues. Reduced hydration demonstrates activation of fibroblasts in the keratinocyte-fibroblast co-culture. In contrast, knockdown of S100A8 or S100A9 by RNA interference in keratinocytes failed to activate fibroblasts. Pretreatment with pharmacological blockers of S100A8/A9 receptors, Toll-like receptor 4 and receptor for advanced glycation end products, inhibits fibroblast activation induced by recombinant S100A8/A9 proteins. Moreover, we observe that local delivery of S100A8 protein results in a marked increase in hypertrophic scarring in the in vivo rabbit ear scar model. Our results indicate that hydration status promotes fibroblast activation and fibrosis by directly affecting the expression of inflammatory signaling in keratinocytes, thereby strongly suggesting S100A8/A9 to be novel targets in preventing scarring.


Asunto(s)
Calgranulina A/biosíntesis , Calgranulina B/biosíntesis , Cicatriz/metabolismo , Epidermis/patología , Fibroblastos/metabolismo , Queratinocitos/metabolismo , Adulto , Animales , Western Blotting , Técnicas de Cocultivo , Dermis/patología , Femenino , Fibrosis/patología , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Masculino , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Agua , Adulto Joven
8.
Exp Dermatol ; 25(8): 604-10, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26997546

RESUMEN

Hypertrophic scarring is a common dermal fibroproliferative disorder characterized by excessive collagen deposition. Prostaglandin E2 (PGE2 ), an important inflammatory product synthesized via the arachidonic acid cascade, has been shown to act as a fibroblast modulator and to possess antifibroblastic activity. However, the mechanism underlying the antifibrotic effect of PGE2 remains unclear. In this study, we explored the effects of PGE2 on TGF-ß1-treated dermal fibroblasts in terms of collagen production and to determine the regulatory pathways involved, as well as understand the antiscarring function of PGE2 in vivo. We found that PGE2 inhibited TGF-ß1-induced collagen synthesis by regulating the balance of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase (TIMP). It did so by upregulating cAMP through the E prostanoid (EP)2 receptor. We determined that inhibition of the TGF-ß1/Smad pathway by PGE2 is associated with its ability to inhibit collagen synthesis. An in vivo study further confirmed that PGE2 inhibits hypertrophic scar formation by decreasing collagen production. Our results demonstrate that the novel anti-scarring function of PGE2 is achieved by balancing MMPs/TIMP expression and decreasing collagen production.


Asunto(s)
Cicatriz/prevención & control , Colágeno/biosíntesis , Dinoprostona/uso terapéutico , Fibroblastos/metabolismo , Animales , Azetidinas , Colforsina , AMP Cíclico/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Evaluación Preclínica de Medicamentos , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Isoindoles , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Conejos , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Sulfonamidas , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta1
9.
J Cell Physiol ; 230(8): 1895-905, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25546411

RESUMEN

Endothelial dysfunction is a major characteristic of diabetic vasculopathy. Protection of the vascular endothelium is an essential aspect of preventing and treating diabetic vascular complications. Although Angiopoietin-1 (Ang-1) is an important endothelial-specific protective factor, whether Ang-1 protects vascular cells undergoing advanced glycation end product (AGE) injury has not been investigated. The aim of the present study was to determine the potential effects of Ang-1 on endothelial cells after exposure to AGE. We show here that Ang-1 prevented AGE-induced vascular leakage by enhancing the adherens junctions between endothelial cells, and this process was mediated by the phosphorylation and membrane localization of VE-cadherin. Furthermore, Ang-1 also protected endothelial cells from AGE-induced death by regulating phosphatidylinositol 3-kinase (PI3K)/Akt-dependent Bad phosphorylation. Our findings suggest that the novel protective mechanisms of Ang-1 on endothelium are achieved by strengthening endothelial cell junctions and reducing endothelial cell death after AGE injury.


Asunto(s)
Angiopoyetina 1/metabolismo , Apoptosis/fisiología , Células Endoteliales/patología , Productos Finales de Glicación Avanzada/toxicidad , Uniones Intercelulares/metabolismo , Angiopoyetina 1/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Línea Celular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Uniones Intercelulares/efectos de los fármacos
10.
EBioMedicine ; 106: 105258, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39068733

RESUMEN

BACKGROUND: Severe burn wounds face two primary challenges: dysregulated cellular impairment functions following infection and an unbalanced wound hydration microenvironment leading to excessive inflammation and collagen deposition. These results in hypertrophic scar contraction, causing significant deformity and disability in survivors. METHODS: A three-dimensional (3D) printed double-layer hydrogel (DLH) was designed and fabricated to address the problem of scar formation after burn injury. DLH was developed using methacrylated silk fibroin (SFMA) and gelatin methacryloyl (GelMA) for the upper layer, and GelMA and hyaluronic acid methacryloyl (HAMA) for the lower layer. To combat infection, copper-epigallocatechin gallate (Cu-EGCG) was incorporated into the lower layer bioink, collectively referred to as DLS. To balance wound hydration levels, HaCaT cells were additionally encapsulated in the upper layer, designed as DLS/c. FINDINGS: DLH demonstrated suitable porosity, appropriate mechanical properties, and excellent biocompatibility. DLS exhibited potent antimicrobial properties, exerted anti-inflammatory effects by regulating macrophage polarisation, and may enhance angiogenesis through the HIF-1α/VEGF pathway. In the DLS/c group, animal studies showed significant improvements in epidermal formation, barrier function, and epidermal hydration, accompanied by reduced inflammation. In addition, Masson's trichrome and Sirius red staining revealed that the structure and ratio of dermal collagen in DLS/c resembled that of normal skin, indicating considerable potential for scarless wound healing. INTERPRETATION: This biomimetic matrix shows promise in addressing the challenges of burn wounds and aiming for scarless repair, with benefits such as anti-infection, epidermal hydration, biological induction, and optimised topological properties. FUNDING: Shown in Acknowledgements.


Asunto(s)
Quemaduras , Impresión Tridimensional , Piel Artificial , Cicatrización de Heridas , Quemaduras/tratamiento farmacológico , Quemaduras/metabolismo , Quemaduras/patología , Cicatrización de Heridas/efectos de los fármacos , Humanos , Animales , Hidrogeles/química , Ratones , Antiinfecciosos/farmacología , Gelatina/química , Cicatriz/patología , Cicatriz/metabolismo , Cicatriz/tratamiento farmacológico , Línea Celular , Fibroínas/química , Ratas , Masculino , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA