Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 51(1): 116, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38227121

RESUMO

BACKGROUND: SOX4 is a transcription factor belonging to the SOX (Sry-related High Mobility Group [HMG] box) family and plays a pivotal role in various biological processes at various stages of life. SOX4 is also expressed in the skin in adults and has been reported to be involved in wound healing, tumor formation, and metastasis. METHODS AND RESULTS: In this study, we investigated the role of SOX4 in keratinocyte phenotypic changes. We generated a SOX4-overexpressing keratinocyte cell line that expresses SOX4 in a doxycycline (DOX)-inducible manner. DOX treatment induced a change from a paving stone-like morphology to a spindle-like morphology under microscopic observation. Comprehensive gene analysis by RNA sequencing revealed increased expression of genes related to anatomical morphogenesis and cell differentiation as well as decreased expression of genes related to epithelial formation and keratinization, suggesting that SOX4 induced EMT-like phenotype in keratinocytes. Differentially expressed genes (DEGs) obtained by RNA-seq were confirmed using qRT-PCR. DOX-treated TY-1 SOX4 showed a decrease in the epithelial markers (KRT15, KRT13, KRT5, and CLDN1) and an increase in the mesenchymal marker FN1. Protein expression changes by Western blotting also showed a decrease in the epithelial marker proteins keratin 15, keratin 13, and claudin 1, and an increase in the mesenchymal marker fibronectin. Removal of DOX from DOX-treated cells also restored the epithelial and mesenchymal markers altered by SOX4. CONCLUSION: Our results indicate that SOX4 reversibly induces an EMT-like phenotype in human keratinocytes via suppression of epithelial marker genes.


Assuntos
Queratinócitos , Fatores de Transcrição SOXC , Pele , Humanos , Western Blotting , Doxiciclina , Expressão Gênica , Fenótipo , Fatores de Transcrição SOXC/genética
2.
Exp Ther Med ; 22(4): 1124, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34466140

RESUMO

Epithelial-mesenchymal transition (EMT) is a cellular process in which epithelial cells lose their epithelial traits and shift to the mesenchymal phenotype, and is associated with various biological events, such as embryogenesis, wound healing and cancer progression. The transcriptional program that promotes phenotype switching is dynamically controlled by transcription factors during EMT, including Snail (SNAI1), twist family bHLH transcription factor (TWIST) and zinc finger E-box binding homeobox 1 (ZEB1). The present study aimed to investigate the molecular mechanisms underlying EMT in squamous epithelial cells. Western blot analysis and immunocytochemical staining identified Slug (SNAI2) as a transcription factor that is induced during transforming growth factor (TGF)-ß1-mediated EMT in the human keratinocyte cell line HaCaT. The effect of SNAI2 overexpression and knockdown on the phenotypic characteristics of HaCaT cells was evaluated. Filamentous actin staining and western blot analysis revealed that the overexpression of SNAI2 did not induce the observed EMT-related phenotypic changes. In addition, SNAI2 knockdown demonstrated almost no impact on the EMT phenotypes induced by TGF-ß1. Notably, DNA microarray analysis followed by comprehensive bioinformatics analysis revealed that the differentially expressed genes upregulated by TGF-ß1 were significantly enriched in cell adhesion and extracellular matrix binding, whereas the genes downregulated in response to TGF-ß1 were significantly enriched in the cell cycle. No enriched gene ontology term and biological pathways were identified in the differentially expressed gene sets of SNAI2-overexpressing cells. In addition, the candidates for master transcription factors regulating the TGF-ß1-induced EMT were identified using transcription factor enrichment analysis. In conclusion, the results of study demonstrated that SNAI2 does not play an essential role in the EMT of HaCaT cells and identified candidate transcription factors that may be involved in EMT-related gene expression induced by TGF-ß1. These findings may enhance the understanding of molecular events in EMT and contribute to the development of a novel therapeutic approach against EMT in cancers and wound healing.

3.
Heliyon ; 7(1): e06102, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33553759

RESUMO

The transient receptor potential melastatin 5 (TRPM5) channel is a monovalent-permeable cation channel that is activated by intracellular Ca2+. Expression of TRPM5 has been shown in taste cells, pancreas, brainstem and olfactory epithelium, and this channel is thought to be involved in controlling membrane potentials. In whole-cell patch-clamp recordings, TRPM5 exhibited voltage-dependent inactivation at negative membrane potentials and time constant of voltage-dependent inactivation of TRPM5 did not depend on the intracellular Ca2+ concentrations between 100 and 500 nM. Alanine substitution at Y913 and I916 in the pore helix of TRPM5 increased time constant of voltage-dependent inactivation. Meanwhile, voltage-dependent inactivation was reduced in TRPM5 mutants having glycine substitution at L901, Y913, Q915 and I916 in the pore helix. From these results, we conclude that the pore helix in the outer pore loop might play a role in voltage-dependent inactivation of TRPM5.

4.
Biomed Rep ; 9(3): 253-258, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30271602

RESUMO

In a previous genome-wide association study, plexin A2 (PLXNA2) was suggested as one of the candidate genes for mandibular prognathism. PLXNA2 encodes plexin A2, a member of the plexin-A family of semaphorin co-receptors. Semaphorin 3A (sema3A) exerts an osteoprotective effect. However, to the best of our knowledge, there have been no previous studies examining the role of sema3A or plexin A2 on human chondrocytes. The objectives of the present study were to examine the function of sema3A and its receptor, plexin A2, in human chondrocytes. Normal human chondrocytes were cultured in media with either a high (100 ng/ml) or a low (1 ng/ml) concentration of sema3A, or without sema3A as a control. Cells and extracellular matrices were assayed for concentrations of protein and parathyroid hormone-related peptide receptor 1 (PTH-R1) using a bicinchoninic acid assay and an enzyme immunoassay, respectively. At culture day 7, the high and low concentrations of exogenous sema3A significantly increased the protein content compared with the control (P=0.0008 and 0.00002, respectively). At culture day 14, a high concentration of exogenous sema3A significantly increased the protein content and decreased the concentration of PTH-R1 compared with the control (P=0.002). The present study revealed novel results that exogenous sema3A suppresses the expression of PTH-R1 in human proliferative chondrocytes and suggested that sema3A may affect human chondrocytes via its receptor, plexin A2.

5.
J Dermatol Sci ; 90(3): 332-342, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29610016

RESUMO

BACKGROUND: Keratinocytes release several factors that are involved in wound contracture and scar formation. We previously reported that a three-dimensional reconstruction model derived from rat skin represents a good wound healing model. OBJECTIVE: We characterized the role of transient receptor potential (TRP) channels in the release of transforming growth factor (TGF)-ß1 from keratinocytes and the differentiation of fibroblasts to identify possible promising pharmacological approaches to prevent scar formation and contractures. METHODS: The three-dimensional culture model was made from rat keratinocytes seeded on a collagen gel in which dermal fibroblasts had been embedded. RESULTS: Among the TRP channel inhibitors tested, the TRPV2 inhibitors SKF96365 and tranilast attenuated most potently keratinocyte-dependent and - independent collagen gel contraction due to TGF-ß signaling as well as TGF-ß1 release from keratinocytes and α-smooth muscle actin production in myofibroblasts. Besides the low amounts detected in normal dermis, TRPV2 mRNA and protein levels were increased after fibroblasts were embedded in the gel. TRPV2 was also expressed in the epidermis and keratinocyte layers of the model. Both inhibitors and TRPV2 siRNA attenuated the intracellular increase of Ca2+ induced by the TRPV agonist 2-aminoethoxydiphenyl borate in TGF-ß1-pretreated fibroblasts. CONCLUSION: This is the first study to show that compounds targeting TRPV2 channels ameliorate wound contraction through the inhibition of TGF-ß1 release and the differentiation of dermal fibroblasts in a culture model.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Miofibroblastos/fisiologia , Canais de Cátion TRPV/antagonistas & inibidores , Fator de Crescimento Transformador beta1/metabolismo , Cicatrização/efeitos dos fármacos , Actinas/metabolismo , Animais , Compostos de Boro/farmacologia , Células Cultivadas , Células Epidérmicas , Epiderme/efeitos dos fármacos , Epiderme/fisiologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Miofibroblastos/efeitos dos fármacos , Cultura Primária de Células , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
6.
Biochem Biophys Res Commun ; 496(2): 381-386, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29326042

RESUMO

Epithelial-mesenchymal transition (EMT) is a biological event in which epithelial cells lose their polarity and cell-cell adhesions and concomitantly acquire mesenchymal traits, and is thought to play an important role in pathological processes such as wound healing and cancer progression. In this study, we evaluated transforming growth factor (TGF)-ß1-treated human keratinocyte HaCaT cells as an in vitro model of EMT. HaCaT cells were changed into an elongated fibroblast-like morphology, which is indicative of EMT in response to TGF-ß1. Phalloidin staining demonstrated the formation of actin stress fibers in TGF-ß1-treated cells. Quantitative RT-PCR analysis revealed that TGF-ß1 increased the mRNA levels of EMT transcription factors (SNAI2, TWIST1, and ZEB1) and mesenchymal markers (CDH2, VIM, and FN1), while it decreased the transcripts of epithelial phenotypic genes (CLDN1, OCLN, KRT5, KRT15, KRT13, and TGM1). Furthermore, we found that KRT13 was drastically suppressed through the reduction of RNA polymerase II occupancy of its promoter, which was accompanied by a decrease in active histone marks (H3K4me3 and H3K27ac) and an increase in a repressive mark (H3K27me3) during EMT. These findings indicate that the TGF-ß1-induced EMT program regulates a subset of epithelial and mesenchymal marker genes, and that KRT13 is transcriptionally suppressed through the modulation of the chromatin state at the KRT13 promoter in HaCaT cells.


Assuntos
Epigênese Genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Queratina-13/genética , Queratinócitos/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Actinas/genética , Actinas/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Citocinas/genética , Citocinas/metabolismo , Transição Epitelial-Mesenquimal/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas , Histonas/genética , Histonas/metabolismo , Humanos , Queratina-13/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Transdução de Sinais , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
7.
J Biol Chem ; 291(16): 8848-61, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929406

RESUMO

FoxO1 binds to insulin response elements located in the promoters of insulin-like growth factor-binding protein 1 (IGFBP1) and glucose-6-phosphatase (G6Pase), activating their expression. Insulin-mediated phosphorylation of FoxO1 promotes cytoplasmic translocation, inhibiting FoxO1-mediated transactivation. We have previously demonstrated that FoxO1 opens and remodels chromatin assembled from the IGFBP1 promoter via a highly conserved winged helix motif. This finding, which established FoxO1 as a "pioneer" factor, suggested a model whereby FoxO1 chromatin remodeling at regulatory targets facilitates binding and recruitment of additional regulatory factors. However, the impact of FoxO1 phosphorylation on its ability to bind chromatin and the effect of FoxO1 loss on recruitment of neighboring transcription factors at its regulatory targets in liver chromatin is unknown. In this study, we demonstrate that an amino acid substitution that mimics insulin-mediated phosphorylation of a serine in the winged helix DNA binding motif curtails FoxO1 nucleosome binding. We also demonstrate that shRNA-mediated loss of FoxO1 binding to the IGFBP1 and G6Pase promoters in HepG2 cells significantly reduces binding of RNA polymerase II and the pioneer factors FoxA1/A2. Knockdown of FoxA1 similarly reduced binding of RNA polymerase II and FoxO1. Reduction in acetylation of histone H3 Lys-27 accompanies loss of FoxO1 and FoxA1/A2 binding. Interdependent binding of FoxO1 and FoxA1/A2 possibly entails cooperative binding because FoxO1 and FoxA1/A2 facilitate one another's binding to IGFPB1 promoter DNA. These results illustrate how transcription factors can nucleate transcriptional events in chromatin in response to signaling events and suggest a model for regulation of hepatic glucose metabolism through interdependent FoxO/FoxA binding.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Insulina/metabolismo , Regiões Promotoras Genéticas/fisiologia , Ativação Transcricional/fisiologia , Animais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Células Hep G2 , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/genética , Humanos , Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Camundongos , Fosforilação/fisiologia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo
8.
Tissue Eng Regen Med ; 13(5): 527-537, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30603433

RESUMO

Three-dimensional (3D) cultures are known to promote cell differentiation. Previously, we investigated the differentiation of rat dermal fibroblasts to α-smooth muscle actin (α-SMA)-positive myofibroblasts through transforming growth factor (TGF)-ß production using a 3D culture model. Here, we investigated the phenotypic change from dermal mesenchymal cells (mostly fibroblasts) to osteoblast-like cells, being inspired by the roles of smooth muscle cells or fibroblasts during vascular calcification. Spindle-shaped cells that grew in heterologous populations out of dermal explants from 2-day-old Wistar rats were cultured within a collagen matrix. α-SMA and alkaline phosphatase (ALP) meßsenger RNA (mRNA) levels initially increased, followed by a rise in Runx2 and osteocalcin (OCN) mRNA levels without calcification. Calcium deposits were produced in the presence of a high concentration of inorganic phosphate (2.1 mM) or ß-glycerophosphate (ßGP, 10 mM) after 2 weeks of culture, and both were sensitive to an inhibitor of type III phosphate transporters. An ALP inhibitor decreased only ßGP-induced calcification. Inhibition of TGF-ß type-I receptors attenuated ALP mRNA levels and ßGP-induced calcification, suggesting that endogenous TGF-ß stimulates ALP activity and then ßGP breakdown. An increase in the number of cells embedded in the collagen gel enhanced the mRNA levels of Runx2 and OCN, but not of ALP. Collectively, several factors are likely to promote the differentiation of dermal mesenchymal cells into osteoblast-like cells and ectopic calcification in a 3D collagen matrix, implying the utility of these cells as a potential autologous cell source for tissue engineering.

9.
Biochem Biophys Res Commun ; 468(1-2): 269-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26505792

RESUMO

In tumor tissues, alterations of gene expression caused by aberrant epigenetic modifications confer phenotypic diversity on malignant cells. Although 3-deazaneplanocin A (DZNep) has been shown to reactivate tumor suppressor genes in several cancer cells, it remains unclear whether DZNep attenuates the malignant phenotypes of oral squamous cell carcinoma (OSCC) cells. In this study, we investigated the effect of DZNep on the expression of genes related to aggressive phenotypes, such as epithelial-mesenchymal transition, in OSCC cells. We found that DZNep reduced the cellular levels of polycomb group proteins (EZH2, SUZ12, BMI1, and RING1A) and the associated trimethylation of Lys27 on histone H3 and monoubiquitination of Lys119 on histone H2A in the poorly differentiated OSCC cell line SAS. Immunocytochemical staining demonstrated that DZNep induced the reorganization of filamentous actin and the membrane localization of E-cadherin associated with cell-cell adhesions. We also found an inhibitory effect of DZNep on cell proliferation using a WST assay. Finally, quantitative RT-PCR analysis demonstrated that genes involved in the aggressive phenotypes (TWIST2, EGFR, ACTA2, TGFB1, WNT5B, and APLIN) were down-regulated, whereas epithelial phenotype genes (CDH1, CLDN4, IVL, and TGM1) were up-regulated in SAS cells treated with DZNep. Collectively, our findings suggest that DZNep reverses the aggressive characteristics of OSCC cells through the dynamic regulation of epithelial plasticity via the reprogramming of gene expression patterns.


Assuntos
Adenosina/análogos & derivados , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Bucais/tratamento farmacológico , Adenosina/farmacologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Histonas/análise , Humanos , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Proteínas do Grupo Polycomb/análise
10.
J Dermatol Sci ; 78(3): 189-96, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25828855

RESUMO

BACKGROUND: CLCA was postulated to be a calcium-activated chloride channel accessory protein. Recent reports indicate that CLCA isoforms are likely to be expressed in different layers of the stratified epithelium of the skin. OBJECTIVE: The present study investigated the transcriptional mechanism by which murine CLCA2 (mCLCA2) is expressed in the transformed keratinocyte line Pam212 that can differentiate. METHODS: A luciferase reporter assay, chromatin immunoprecipitation (ChIP) assay, reverse transcription-PCR, and immunocytochemistry were performed using Pam212 cells. RESULTS: Promoter activity of mCLCA2 was inhibited profoundly by site-directed mutagenesis of a putative nuclear factor-κB (NF-κB) binding site and by treatment with siRNA against p65. ChIP and transcription factor assays showed the specific association of endogenously activated p65 protein with the NF-κB binding domain. As confirmed by the nuclear translocation of p65, tumor necrosis factor α and caffeic acid phenethyl ester (CAPE) increased and decreased mCLCA2 promoter activity, respectively, but exhibited modest effects on endogenous mCLCA2 expression in cells in culture medium containing 0.05 mM Ca(2+). When the Ca(2+) concentration was raised to 1.0mM, the mRNA and protein levels of mCLCA2 increased as well as those of the differentiation markers keratin 1 (K1) and K10. CAPE profoundly suppressed only the Ca(2+)-triggered expression of mCLCA2, not K1 or K10. Immunohistochemistry of native skin and organotypic 3D cultures confirmed the distribution of the CLCA2 homolog in differentiated cells. CONCLUSION: The present study revealed for the first time that basal NF-κB activity is involved in the Ca(2+)-dependent regulation of mCLCA2 expression in a mouse keratinocyte line.


Assuntos
Canais de Cloreto/genética , Queratinócitos/metabolismo , NF-kappa B/fisiologia , Transcrição Gênica , Animais , Cálcio/metabolismo , Células Cultivadas , Canais de Cloreto/fisiologia , Regulação da Expressão Gênica , Camundongos , Regiões Promotoras Genéticas
11.
Biochem Biophys Res Commun ; 458(1): 161-7, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25637531

RESUMO

The directional migration of epithelial cells is crucial for wound healing. Among integrins, a family of cell adhesion receptors, integrin ß4 has been assumed to be a promigratory factor, in addition to its role in stable adhesion. In turn, Ca(2+) signaling is also a key coordinator of migration. Keratinocytes reportedly express transient receptor potential vanilloid channels (TRPV1); however, the function of these channels as a regulator of intracellular Ca(2+) level in cell migration has remained uncharacterized. In the present study, we investigated the role of TRPV1 in directional migration related to integrin ß4 using a scratch wound assay on a confluent monolayer sheet of murine keratinocytes (Pam212 cells). Double immunofluorescence staining revealed the de novo expression of integrin ß4 and TRPV1 in migrating cells at the wound edge in response to scratch wounding, and both expression levels were almost matched. Epidermal growth factor (EGF) not only promoted keratinocyte migration, but also caused the further up-regulation of both integrin ß4 and TRPV1. In addition, the knockdown of the integrin ß4 or TRPV1 gene significantly impeded wound closure. The TRPV1 agonist capsaicin significantly promoted migration, while a selective TRPV1 antagonist inhibited it. The gene knockdown of TRPV1 inhibited the expression of the integrin ß4 gene and that of ß4 protein in migrating cells. These findings suggest that TRPV1 may stimulate directional migration directly by eliciting a Ca(2+) signal or indirectly via integrin ß4 expression.


Assuntos
Integrina alfa6beta4/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Capsaicina/farmacologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cobalto/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Inativação Gênica , Integrina alfa6beta4/genética , Queratinócitos/efeitos dos fármacos , Camundongos , Canais de Cátion TRPV/genética , Regulação para Cima , Cicatrização
12.
BMC Cancer ; 14: 988, 2014 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-25527207

RESUMO

BACKGROUND: Epigenetic modifications play important roles in the regulation of gene expression determining cellular phenotype as well as various pathologies such as cancer. Although the loss of keratin 13 (KRT13) is reportedly linked to malignant transformation of oral epithelial cells, the molecular mechanisms through which KRT13 is repressed in oral squamous cell carcinoma (OSCC) remain unclear. The aim of this study is to identify the epigenetic alterations of the KRT13 gene in OSCCs. METHODS: We investigated KRT13 expression levels and chromatin modifications of the KRT13 promoter in the three OSCC cell lines (HSC4, HSC3, and SAS). The expression levels of KRT13 protein and mRNA were analyzed by western blotting and quantitative reverse-transcription polymerase chain reaction, respectively, and the localization of KRT13 protein was detected by immunofluorescence. DNA methylation and histone modifications in the KRT13 promoter were determined by bisulfite sequencing and chromatin immunoprecipitation (ChIP), respectively. For the pharmacological depletion of Polycomb repressive complex 2 (PRC2), cells were treated with 3-deazaneplanocin A (DZNep). RESULTS: KRT13 expression was transcriptionally silenced in the HSC3 and SAS cells and post-transcriptionally repressed in the HSC4 cells, while the KRT13 promoter was hypermethylated in all of the three OSCC cell lines. ChIP analysis revealed that PRC2-mediated trimethylation of Lys 27 on histone H3 (H3K27me3) was increased in the KRT13 promoter in the HSC3 and SAS cells. Finally, we demonstrated that the treatment of SAS cells with DZNep reactivated the transcription of KRT13 gene. CONCLUSIONS: Our data provide mechanistic insights into the epigenetic silencing of KRT13 genes in OSCC cells and might be useful for the development of diagnostic markers and novel therapeutic approaches against OSCCs.


Assuntos
Carcinoma de Células Escamosas/genética , Epigênese Genética , Queratina-13/genética , Neoplasias Bucais/genética , Adenosina/análogos & derivados , Adenosina/farmacologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Queratina-13/metabolismo , Metilação , Neoplasias Bucais/metabolismo , Regiões Promotoras Genéticas , Ativação Transcricional/efeitos dos fármacos
13.
J Pharmacol Sci ; 124(2): 230-43, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24492413

RESUMO

Transforming growth factor-ß1 (TGF-ß1) reportedly causes the differentiation of fibroblasts to myofibroblasts during wound healing. We investigated the mechanism underlying the activation of latent TGF-ß1 released by keratinocytes in efforts to identify promising pharmacological approaches for the prevention of hypertrophic scar formation. A three-dimensional collagen gel matrix culture was prepared using rat keratinocytes and dermal fibroblasts. Stratified keratinocytes promoted the TGF receptor-dependent increase in α-smooth muscle actin (α-SMA) immunostaining and mRNA levels in fibroblasts. Latent TGF-ß1 was found to be localized suprabasally and secreted. α-SMA expression was inhibited by an anti-αv-integrin antibody and a matrix metalloproteinase (MMP) inhibitor, GM6001. In a two-dimensional fibroblast culture, α-SMA expression depended on the production of endogenous TGF-ß1 and required αv-integrin or MMP for the response to recombinant latent TGF-ß1. In keratinocyte-conditioned medium, MMP-dependent latent TGF-ß1 secretion was detected. Applying this medium to the fibroblast culture enhanced α-SMA production. This effect was decreased by GM6001, the anti-αv-integrin antibody, or the preabsorption of latent TGF-ß1. These results indicate that keratinocytes secrete latent TGF-ß1, which is liberated to fibroblasts over distance and is activated to produce α-SMA with the aid of a positive-feedback loop. MMP inhibition was effective for targeting both keratinocytes and fibroblasts in this model.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fibroblastos/citologia , Queratinócitos/metabolismo , Pele/citologia , Fator de Crescimento Transformador beta1/farmacologia , Cicatrização/fisiologia , Actinas/metabolismo , Animais , Anticorpos/farmacologia , Células Cultivadas , Cicatriz/genética , Cicatriz/patologia , Cicatriz/prevenção & controle , Dipeptídeos/farmacologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Integrina alfa5/imunologia , Inibidores de Metaloproteinases de Matriz/farmacologia , Miofibroblastos/patologia , Ratos , Fator de Crescimento Transformador beta1/metabolismo
14.
J Biol Chem ; 288(7): 4831-43, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23297403

RESUMO

We previously found that a rat CLCA homologue (rCLCA-f) modulates Ca(2+)-dependent Cl(-) transport in the ductal cells of the rat submandibular gland. CLCA proteins have been shown to be multifunctional, with roles in, for example, cell adhesion. Here, we describe the mRNA and protein expressions of a splicing isoform of rat rCLCA (rCLCA-t). This isoform is a 514-amino acid protein containing a C-terminal 59-amino acid that is distinct from the rCLCA-f sequence. Immunohistochemistry revealed rCLCA-t to be located in the basal cells of the rat submandibular gland excretory duct and the stratum basale of rat epidermis, whereas rCLCA-f was detected in cells during the process of differentiation. In a heterologous expression system, rCLCA-t was found to be a membrane protein present predominantly in the perinuclear region, and not to be either present on the cell surface or secreted. rCLCA-t failed to enhance ionomycin-induced Cl(-) conductance (unlike rCLCA-f). When compared with rCLCA-f, it weakened cell attachment to a greater extent and in a manner that was evidently modulated by intracellular Ca(2+), protein kinase C, and ß(1)-integrin. rCLCA-t was found to associate with RACK1 (receptor for activated C kinase) and to reduce expression of mature ß(1)-integrin. Treatment of rat skin with rCLCA-t siRNA increased the expression of ß(1)-integrin in the stratum basale of the epidermis. These results are consistent with cell-specific splicing of rCLCA mRNA playing a role in the modulation of the adhesive potential of undifferentiated epithelial cells.


Assuntos
Canais de Cloreto/fisiologia , Células Epiteliais/citologia , Proteínas de Ligação ao GTP/metabolismo , Integrina beta1/metabolismo , Processamento Alternativo , Animais , Adesão Celular , Diferenciação Celular , Canais de Cloreto/química , Canais de Cloreto/metabolismo , Epiderme/metabolismo , Masculino , Modelos Genéticos , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Ligação Proteica , Isoformas de Proteínas , Ratos , Ratos Wistar , Receptores de Quinase C Ativada
15.
J Biol Chem ; 287(27): 23236-45, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22589549

RESUMO

Mutations of the HNF4A gene cause a form of maturity-onset diabetes of the young (MODY1) that is characterized by impairment of pancreatic ß-cell function. HNF4α is a transcription factor belonging to the nuclear receptor superfamily (NR2A1), but its target genes in pancreatic ß-cells are largely unknown. Here, we report that ankyrin repeat and sterile α motif domain containing 4b (Anks4b) is a target of HNF4α in pancreatic ß-cells. Expression of Anks4b was decreased in both ßHNF4α KO islets and HNF4α knockdown MIN6 ß-cells, and HNF4α activated Anks4b promoter activity. Anks4b bound to glucose-regulated protein 78 (GRP78), a major endoplasmic reticulum (ER) chaperone protein, and overexpression of Anks4b enhanced the ER stress response and ER stress-associated apoptosis of MIN6 cells. Conversely, suppression of Anks4b reduced ß-cell susceptibility to ER stress-induced apoptosis. These results indicate that Anks4b is a HNF4α target gene that regulates ER stress in ß-cells by interacting with GRP78, thus suggesting that HNF4α is involved in maintenance of the ER.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Diabetes Mellitus Tipo 2 , Estresse do Retículo Endoplasmático/fisiologia , Proteínas de Choque Térmico/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Feminino , Perfilação da Expressão Gênica , Células Secretoras de Insulina/citologia , Insulinoma , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Neoplasias Pancreáticas , Proteômica , Ativação Transcricional/fisiologia
16.
Biochem Biophys Res Commun ; 407(3): 620-5, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21426901

RESUMO

KCNQ1, located on 11p15.5, encodes a voltage-gated K(+) channel with six transmembrane regions, and loss-of-function mutations in the KCNQ1 gene cause hereditary long QT syndrome. Recent genetic studies have identified that single nucleotide polymorphisms located in intron 15 of the KCNQ1 gene are strongly associated with type 2 diabetes and impaired insulin secretion. In order to understand the role of KCNQ1 in insulin secretion, we introduced KCNQ1 into the MIN6 mouse ß-cell line using a retrovirus-mediated gene transfer system. In KCNQ1 transferred MIN6 cells, both the density of the KCNQ1 current and the density of the total K(+) current were significantly increased. In addition, insulin secretion by glucose, pyruvate, or tolbutamide was significantly impaired by KCNQ1-overexpressing MIN6 cells. These results suggest that increased KCNQ1 protein expression limits insulin secretion from pancreatic ß-cells by regulating the potassium channel current.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canal de Potássio KCNQ1/metabolismo , Animais , Linhagem Celular , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Secreção de Insulina , Canal de Potássio KCNQ1/genética , Camundongos , Camundongos Endogâmicos C57BL , Retroviridae , Transfecção
17.
Biochem Biophys Res Commun ; 389(1): 133-7, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19715677

RESUMO

Collectrin is a novel target gene of hepatocyte nuclear factor-1alpha in pancreatic beta-cells and controls insulin exocytosis. Although glucose is known to stimulate the expression of genes of the insulin secretory pathway, there is no information on how glucose regulates collectrin expression. We investigated the effects of glucose on the expression of collectrin in MIN6 beta-cell line. Glucose, in a dose-dependent manner, increased collectrin protein levels without changing collectrin mRNA levels and protein stability, indicating that glucose stimulation of collectrin protein expression is primarily mediated at a translational level. Although mannose and pyruvate also increased collectrin protein expression level, neither 2-deoxyglucose, mitochondrial fuels leucine and glutamate, sulphonylurea nor Ca(2+) channel blockers, mimicked the effects of glucose. These data indicate the involvement of mitochondrial TCA cycle intermediates, distal to pyruvate, in the regulation of collectrin protein expression in beta-cells.


Assuntos
Ciclo do Ácido Cítrico , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Glicoproteínas de Membrana/biossíntese , Animais , Linhagem Celular , Glucose/farmacologia , Insulina/biossíntese , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Biossíntese de Proteínas/efeitos dos fármacos
18.
Oncol Rep ; 20(3): 663-8, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18695921

RESUMO

Matrix metalloproteinases (MMPs) play important roles in the invasion and metastasis to soft tissues of carcinomas including, oral squamous cell carcinomas (SCCs). Although, osteoclastic bone resorption is an important step in bone involvement in a variety of malignancies, the mechanism of bone involvement of oral SCC remains unclear. Once cancer cells arrest in bone, the bone is a storehouse of a variety of cytokines and growth factors and thus provides an extremely fertile environment for cell growth. The bone-invasive oral cancer cell line, BHY, transcriptionally expressed detectable levels of TGF-beta, IL-1beta, IL-8, parathyroid hormone-related protein (PTHrP) and vascular endothelial growth factor (VEGF) mRNAs and failed to express GM-CSF, IL-6, and TNF-alpha. Furthermore, the BHY-conditioned medium greatly upregulated IL-6 and RANKL/ODF mRNA expression in osteoblasts, suggesting a potential indirect stimulation of osteoclastogenesis via the osteogenic lineage. Seven out of eleven patients with carcinomas of the lower alveolus and gingiva showing infiltrative bone involvement expressed PTHrP mRNA. These data suggest that the occurrence of PTHrP may be an indication of developing oral malignant carcinomas.


Assuntos
Carcinoma de Células Escamosas/patologia , Diferenciação Celular , Neoplasias Bucais/patologia , Osteoblastos/citologia , Osteoclastos/citologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Animais , Células da Medula Óssea , Carcinoma de Células Escamosas/metabolismo , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Citocinas/metabolismo , Expressão Gênica , Neoplasias Gengivais/genética , Neoplasias Gengivais/metabolismo , Neoplasias Gengivais/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Bucais/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
J Biol Chem ; 282(49): 35583-93, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17923482

RESUMO

FoxO1, a member of the forkhead rabdomyosarcoma (FoxO) subfamily of transcription factors, binds DNA via a highly conserved winged-helix "forkhead box" motif used by other regulatory proteins to mediate their effects through chromatin binding and remodeling. To examine how FoxO1 regulates target genes in chromatin, we studied the binding of purified recombinant FoxO1 protein to nucleosome particles and chromatin arrays containing the insulin-like growth factor-binding protein 1 promoter. We found that FoxO1 is able to bind to its cognate sites within the insulin-like growth factor-binding protein 1 promoter on a nucleosome. This binding stably perturbs core histone:DNA contacts extending up- and downstream from sites of FoxO1 binding without disrupting the underlying core particle. FoxO1 is able to harness these capabilities to bind to and de-condense linker histone-compacted chromatin arrays. Chromatin opening by FoxO1 requires both the N and C termini of the protein, which are also required for high affinity core histone binding and, in the case of the N terminus, nucleosome perturbation. We suggest that the chromatin binding and remodeling functions revealed here for FoxO1 endow all FoxO factors with the ability to initiate and dynamically modulate active chromatin states, enabling their diverse roles as gene regulatory factors in metabolism, cell survival, apoptosis, cell cycle progression, DNA repair, and protection against oxidative stress.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , DNA/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Sobrevivência Celular/fisiologia , Sistema Livre de Células/metabolismo , DNA/genética , Reparo do DNA/fisiologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Histonas/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Camundongos , Nucleossomos/genética , Estresse Oxidativo/fisiologia , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/fisiologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
20.
Biochem Biophys Res Commun ; 355(2): 438-43, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17306229

RESUMO

E1AF was first identified as a transcription factor that binds to enhancer motifs of the adenovirus E1A gene and is thought to be a human homologue of mouse PEA3, one of the ets oncoprotein families. Here we show the effect of E1A on the gene expression and function of E1AF. E1A repressed the activity of E1AF promoter, and the N-terminal region of E1A, which is involved in the oncogenic activity of E1A, was essential for this repression. The ability as a transcription factor of E1AF, as well as those of the other PEA3 subfamily members ER81 and ERM, was also repressed by E1A via the same oncogenic domain. Furthermore, E1AF repressed the transformation activity of E1A cooperating with E1B, whereas the other ets family Ets-1 enhanced this activity. These results suggest that E1AF is one of the targets of E1A.


Assuntos
Proteínas E1A de Adenovirus/fisiologia , Regulação da Expressão Gênica/genética , Proteína Proto-Oncogênica c-ets-1/genética , Proteínas E1A de Adenovirus/química , Proteínas E1A de Adenovirus/genética , Animais , Linhagem Celular , Humanos , Luciferases/genética , Plasmídeos , Regiões Promotoras Genéticas , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...