Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nephrology (Carlton) ; 27(5): 458-466, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35213087

RESUMO

AIM: Tenascin-C (TNC), a non-structural extracellular matrix glycoprotein, is transiently expressed during development or after injury, playing an important role in injury and repair process. The potential role of TNC in the pathogenesis of IgA nephropathy (IgAN) remains to be clarified. METHODS: Immunohistochemistry staining for TNC was conducted on paraffin-embedded slices from renal biopsies of 107 IgAN patients, and correlation analysis was made between mesangial TNC expression and clinic-pathological parameters. In situ hybridization for TNC mRNA was further performed to figure out the cells that express TNC within glomeruli. In vitro experiments were also carried out on mouse mesangial cells (SV40 MES13) to elucidate the effect of TNC on mesangial cells. RESULTS: TNC was expressed in the mesangial area of IgAN, as well as in fibrotic regions. Correlation analysis showed that higher mesangial TNC was associated with higher level of proteinuria, lower estimated glomerular filtration rate and more serious pathological lesions (MEST score). In situ hybridization revealed that abundant TNC mRNA expression was observed in the affected glomeruli of IgAN, but not in minimal change disease. Moreover, TNC mRNA co-localized with PDGFRß mRNA, but not with PODXL mRNA, suggesting that TNC mRNA was expressed in the mesangial cells within glomeruli in IgAN. In vitro experiments showed that exogenous TNC promoted matrix protein production and mesangial cell proliferation, which was attenuated by an epidermal growth factor receptor inhibitor. CONCLUSION: Taken together, these results suggest that mesangial cell-derived TNC contributes to mesangial matrix expansion and mesangial cell proliferation, which is a potential therapeutic target in IgAN.


Assuntos
Glomerulonefrite por IGA , Células Mesangiais , Animais , Proliferação de Células , Matriz Extracelular/metabolismo , Glomerulonefrite por IGA/patologia , Humanos , Células Mesangiais/patologia , Camundongos , Tenascina/genética , Tenascina/farmacologia
2.
Am J Pathol ; 190(10): 2123-2135, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32650003

RESUMO

Tenascin-C (TNC) is strongly expressed by fibroblasts and cancer cells in breast cancer. To assess the effects of TNC on stromal formation, we examined phenotypic changes in human mammary fibroblasts treated with TNC. The addition of TNC significantly up-regulated α-smooth muscle actin (α-SMA) and calponin. TNC increased the number of α-SMA- and/or calponin-positive cells with well-developed stress fibers in immunofluorescence, which enhanced contractile ability in collagen gel contraction. The treatment with TNC also significantly up-regulated its own synthesis. Double immunofluorescence of human breast cancer tissues showed α-SMA- and/or calponin-positive myofibroblasts in the TNC-deposited stroma. Among several receptors for TNC, the protein levels of the αv and ß1 integrin subunits were significantly increased after the treatment. Immunofluorescence showed the augmented colocalization of αv and ß1 at focal adhesions. Immunoprecipitation using an anti-αv antibody revealed a significant increase in coprecipitated ß1 with TNC in lysates. The knockdown of αv and ß1 suppressed the up-regulation of α-SMA and calponin. The addition of TNC induced the phosphorylation of SMAD2/3, whereas SB-505124 and SIS3 blocked myofibroblast differentiation. Therefore, TNC enhances its own synthesis by forming a positive feedback loop and increases integrin αvß1 heterodimer levels to activate transforming growth factor-ß signaling, which is followed by a change to highly contractile myofibroblasts. TNC may essentially contribute to the stiffer stromal formation characteristic of breast cancer tissues.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos/patologia , Miofibroblastos/patologia , Tenascina/farmacologia , Neoplasias da Mama/metabolismo , Diferenciação Celular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/farmacologia , Fibroblastos/metabolismo , Humanos , Miofibroblastos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/farmacologia , Fosforilação/efeitos dos fármacos , Receptores de Vitronectina/metabolismo , Transdução de Sinais/fisiologia , Tenascina/metabolismo , Fator de Crescimento Transformador beta/metabolismo
3.
Cytotherapy ; 20(8): 1061-1076, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30087008

RESUMO

BACKGROUND: Impaired cutaneous wound healing is common in humans, and treatments are often ineffective. Based on the significant emotional and economic burden of impaired wound healing, innovative therapies are needed. The potential of mesenchymal stromal cell (MSC)-secreted factors to treat cutaneous wounds is an active area of research that is in need of refinement before effective clinical trials can be initiated. The aims of the present study were to (i) study which MSC-secreted factors stimulate dermal fibroblast (DF) migration in vitro and (ii) evaluate the potential of these factors to promote wound healing in vivo. METHODS: To this end, MSCs were isolated from the peripheral blood of healthy horses, a physiologically relevant large animal model appropriate for translational wound-healing studies. Conditioned medium (CM) from cultured equine MSCs was analyzed using liquid chromatography-mass spectrophotometry (LC-MS/MS) to identify secreted proteins of interest. Double-stranded RNA-mediated interference (RNAi) was used to silence the genes encoding selected proteins, and the effects of CM from these transfected MSCs on migration of cultured equine DF cells in vitro and full-thickness wounds in mice were evaluated. RESULTS: We found that MSC-derived plasminogen activator inhibitor-1 (PAI-1) and tenascin-C significantly increased DF migration in vitro and improved wound healing in vivo by decreasing time to wound closure. DISCUSSION: These results suggest that in a complex wound environment, MSC-secreted factors PAI-1 and tenascin-C contribute to the positive effect of therapeutically applied MSC CM on wound healing.


Assuntos
Derme/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Inibidor 1 de Ativador de Plasminogênio , Tenascina , Cicatrização/efeitos dos fármacos , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Derme/citologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiologia , Feminino , Fibroblastos/fisiologia , Cavalos , Células-Tronco Mesenquimais/citologia , Camundongos , Células NIH 3T3 , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Tenascina/metabolismo , Tenascina/farmacologia , Cicatrização/fisiologia
4.
Proc Natl Acad Sci U S A ; 111(8): 3080-5, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24516133

RESUMO

Sphingosine 1-phosphate (S1P) plays a role in lymphocyte egress from lymphoid organs. However, it remains unclear how S1P production and secretion are regulated. We show that under inflammatory conditions, α9 integrin, which is closely associated with activated ß1 integrin, and its ligand, tenascin-C, colocalize on medullary and cortical sinuses of draining lymph nodes (dLNs), which is a gate for lymphocyte exit, and that inhibition of lymphocyte egress is evident by blockade of α9 integrin-mediated signaling at dLNs. Based on in vitro analysis using lymphatic endothelial cells obtained from mice embryos, we suggested the possibility that stimulation of lymphatic endothelial cells by tenascin-C enhances S1P secretion in an α9 integrin-dependent manner without affecting S1P synthesis and/or degradation. Blockade of α9 integrin-mediated signaling reduced lymphocyte egress from dLNs in several models, including experimental autoimmune encephalomyelitis, where it improved clinical scores and pathology. Therefore, manipulating α9 integrin function may offer a therapeutic strategy for treating various inflammatory disorders.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Células Endoteliais/metabolismo , Vigilância Imunológica/imunologia , Cadeias alfa de Integrinas/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Citometria de Fluxo , Adjuvante de Freund , Técnicas Histológicas , Linfonodos/citologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Esfingosina/metabolismo , Estatísticas não Paramétricas , Tenascina/farmacologia
5.
Int J Mol Sci ; 18(1)2017 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-28106752

RESUMO

The extracellular matrix (ECM) molecule tenascin C (TNC) is known to be highly expressed under various pathological conditions such as inflammation and cancer. It has been reported that the expression of TNC is correlated with the malignant potential of cancer. In our laboratory, it was found that the peptide derived from the alternative splicing domain A2 in TNC, termed TNIIIA2, has been shown to influence a variety of cellular processes, such as survival, proliferation, migration, and differentiation. In this study, we investigated the effect of TNC/TNIIIA2 on the invasion and metastasis of colon cancer cells, Colon26-M3.1, or PMF-Ko14, using an in vitro and in vivo experimental system. The degree of cell invasion was increased by the addition of TNC and TNIIIA2 in a dose-dependent manner. The invasion by TNC and TNIIIA2 were suppressed by an MMP inhibitor or TNIIIA2-blocking antibody. In an in vivo experiment, pulmonary metastasis was promoted conspicuously by the addition of TNIIIA2. In this study, we found that colon cancer cell invasion and metastasis was accelerated by TNC/TNIIIA2 via MMP induction. This result suggests the possibility of a new strategy targeting TNC/TNIIIA2 for colon cancer.


Assuntos
Movimento Celular/efeitos dos fármacos , Neoplasias do Colo/enzimologia , Metaloproteinases da Matriz/metabolismo , Peptídeos/farmacologia , Tenascina/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Relação Dose-Resposta a Droga , Proteínas da Matriz Extracelular/química , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Inibidores de Metaloproteinases de Matriz/farmacologia , Metaloproteinases da Matriz/genética , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tenascina/química
6.
Glia ; 64(3): 374-85, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26497118

RESUMO

Peripheral nerve regeneration requires precise coordination and dynamic interaction among various types of cells in the tissue. It remains unclear, however, whether the cellular crosstalk between fibroblasts and Schwann cells (SCs) is related to phenotype modulation of SCs, a critical cellular process after peripheral nerve injury. In this study, microarray analysis revealed that a total of 6,046 genes were differentially expressed in the proximal nerve segment after sciatic nerve transection in rats, and bioinformatics analysis further identified tenascin-C (TNC), an extracellular matrix (ECM) protein, as a key gene regulator. TNC was abundantly produced by nerve fibroblasts accumulating at the lesion site, rather than by SCs as usually expected. TNC significantly promoted SC migration without effects on SC proliferation in primary culture. In co-culture of fibroblasts and SCs, inhibition of TNC expression either by siRNA transfection or antibody blockade could suppress SC migration, while TNC-stimulated SC migration was mediated by TNC binding to ß1-integrin receptor in SCs through activation of Rac1 effectors. The in vivo evidence showed that exogenous TNC protein enhanced SC migration and axonal regrowth. Our results highlight that TNC-mediated cellular interaction between fibroblasts and SCs may regulate SC migration through ß1-integrin-dependent pathway during peripheral nerve regeneration.


Assuntos
Movimento Celular/efeitos dos fármacos , Integrina beta1/metabolismo , Regeneração Nervosa/fisiologia , Células de Schwann/efeitos dos fármacos , Neuropatia Ciática/patologia , Transdução de Sinais/efeitos dos fármacos , Tenascina/farmacologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Fibroblastos/fisiologia , Redes Reguladoras de Genes , Masculino , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Neuropatia Ciática/fisiopatologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Tenascina/metabolismo , Fatores de Tempo , Cicatrização/efeitos dos fármacos , Cicatrização/genética , Proteínas rho de Ligação ao GTP/metabolismo
7.
Mol Vis ; 22: 436-45, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27186070

RESUMO

PURPOSE: We previously demonstrated that tenascin-C was highly expressed in the fibrovascular membranes (FVMs) of patients with proliferative diabetic retinopathy (PDR). However, its role in the pathogenesis of FVMs has not been determined. The purpose of this study was to investigate what role tenascin-C plays in the formation and angiogenesis of FVMs. METHODS: The level of tenascin-C was determined by sandwich enzyme-linked immunosorbent assay in the vitreous samples collected from patients with PDR and with a macular hole as control. The locations of tenascin-C, α- smooth muscle actin (SMA), CD34, glial fibrillary acidic protein (GFAP), and integrin αV in the FVMs from PDR patients were determined by immunohistochemistry. We also measured the in vitro expression of the mRNA and protein of tenascin-C in vascular smooth muscle cells (VSMCs) stimulated by interleukin (IL)-13. The effects of tenascin-C on cell proliferation, migration, and tube formation were determined in human retinal endothelial cells (HRECs) in culture. RESULTS: The mean vitreous levels of tenascin-C were significantly higher in patients with PDR than in patients with a macular hole (p<0.001). Double immunofluorescence analyses of FVMs from PDR patients showed that tenascin-C co-stained FVMs with α-SMA, CD34, and integrin αV but not with GFAP. In addition, IL-13 treatment increased both the expression and secretion of tenascin-C by VSMCs in a dose-dependent manner. Tenascin-C exposure promoted proliferation, migration, and tube formation in HRECs. Tenascin-C neutralizing antibody significantly blocked the tube formation by HRECs exposed to VSMC-IL-13-conditioned medium. CONCLUSIONS: Our findings suggest that tenascin-C is secreted from VSMCs and promotes angiogenesis in the FVMs associated with PDR.


Assuntos
Retinopatia Diabética/metabolismo , Membrana Epirretiniana/metabolismo , Neovascularização Retiniana/metabolismo , Tenascina/fisiologia , Corpo Vítreo/metabolismo , Actinas/metabolismo , Idoso , Antígenos CD34/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Retinopatia Diabética/patologia , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Membrana Epirretiniana/patologia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/metabolismo , RNA Mensageiro/genética , Neovascularização Retiniana/patologia , Perfurações Retinianas/metabolismo , Perfurações Retinianas/patologia , Vasos Retinianos/citologia , Tenascina/farmacologia
8.
Biosci Biotechnol Biochem ; 80(8): 1470-7, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27031437

RESUMO

Tenascin-C (TN-C) is an extracellular matrix glycoprotein markedly upregulated during liver fibrosis. The study is performed to explore the role of TN-C during the growth and activation of hepatic stellate cells (HSCs). We found that TN-C was accumulated accompanying with the HSC activation. Our data on cell migration assay revealed that the rTN-C treatment enhanced HSC migration in a dose- and time-dependent manner, but did not influence their proliferation. HSCs transfected with pTARGET-TN-C overexpression vector displayed increased the type I collagen (Col I) production. TN-C overexpression enhanced the process of HSC activation through TGF-ß1 signaling. Moreover, the anti-α9ß1 integrin antibody treatment blocked the TN-C-driven Col I increase in rat HSCs. Collectively, TN-C had a positive role in activation of HSCs mediated by TGF-ß1 and α9ß1 integrin, manifesting elevation of Col I production and promotion of cell migration. Our results provide a potential insight for the therapy of hepatic fibrosis.


Assuntos
Colágeno Tipo I/genética , Células Estreladas do Fígado/efeitos dos fármacos , Integrinas/genética , Tenascina/farmacologia , Fator de Crescimento Transformador beta1/genética , Animais , Anticorpos/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/genética , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Integrinas/antagonistas & inibidores , Integrinas/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Modelos Biológicos , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Transfecção , Fator de Crescimento Transformador beta1/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(45): 18220-5, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24145401

RESUMO

Achieving an AIDS-free generation will require elimination of postnatal transmission of HIV-1 while maintaining the nutritional and immunologic benefits of breastfeeding for infants in developing regions. Maternal/infant antiretroviral prophylaxis can reduce postnatal HIV-1 transmission, yet toxicities and the development of drug-resistant viral strains may limit the effectiveness of this strategy. Interestingly, in the absence of antiretroviral prophylaxis, greater than 90% of infants exposed to HIV-1 via breastfeeding remain uninfected, despite daily mucosal exposure to the virus for up to 2 y. Moreover, milk of uninfected women inherently neutralizes HIV-1 and prevents virus transmission in animal models, yet the factor(s) responsible for this anti-HIV activity is not well-defined. In this report, we identify a primary HIV-1-neutralizing protein in breast milk, Tenascin-C (TNC). TNC is an extracellular matrix protein important in fetal development and wound healing, yet its antimicrobial properties have not previously been established. Purified TNC captured and neutralized multiclade chronic and transmitted/founder HIV-1 variants, and depletion of TNC abolished the HIV-1-neutralizing activity of milk. TNC bound the HIV-1 Envelope protein at a site that is induced upon engagement of its primary receptor, CD4, and is blocked by V3 loop- (19B and F39F) and chemokine coreceptor binding site-directed (17B) monoclonal antibodies. Our results demonstrate the ability of an innate mucosal host protein found in milk to neutralize HIV-1 via binding to the chemokine coreceptor site, potentially explaining why the majority of HIV-1-exposed breastfed infants are protected against mucosal HIV-1 transmission.


Assuntos
Síndrome da Imunodeficiência Adquirida/transmissão , HIV-1/efeitos dos fármacos , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Leite Humano/química , Tenascina/farmacologia , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Western Blotting , Linhagem Celular , Cromatografia por Troca Iônica , Relação Dose-Resposta a Droga , Feminino , Humanos , Imunoprecipitação , Concentração Inibidora 50 , Espectrometria de Massas , Tenascina/metabolismo , Proteínas do Envelope Viral/metabolismo
10.
J Biol Chem ; 289(25): 17699-708, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24808173

RESUMO

Tenascin-C is an adhesion modulatory matrix protein that is highly expressed in tumors; however, its biochemical activity involved in tumorigenesis is not fully understood. On the other hand, increasing evidence indicates the importance of integrin α5ß1 in cancer development. We previously demonstrated that tenascin-C harbors a functional site that can be released as a proadhesive peptide such as TNIIIA2. Peptide TNIIIA2 is capable of inducing activation of ß1-integrins including α5ß1 via syndecan-4. In this study the proadhesive effect of TNIIIA2 was characterized by potentiated and sustained activation of integrin α5ß1. Based on this effect, TNIIIA2 rendered nontransformed fibroblasts (NIH3T3) resistant to serum deprivation-elicited anoikis through activation of the Akt/Bcl-2 pathway. Moreover, TNIIIA2 hyperstimulated PDGF-dependent proliferation of NIH3T3 by activating integrin α5ß1. Tenascin-C, a parental protein of TNIIIA2, also stimulated PDGF-dependent proliferation, which was blocked by a matrix metalloproteinase-2/9 inhibitor and an anti-TNIIIA2 function-blocking antibody, suggesting proteolytic exposure of the proadhesive effect of TNIIIA2. Mechanistic analyses revealed that TNIIIA2 induced a lateral association of PDGF receptor ß with the molecular complex of activated integrin α5ß1 and syndecan-4 in the membrane microdomains enriched with cholesterol/caveolin-1, resulting in prolonged activation of PDGF receptor ß and the subsequent Ras/mitogen-activated protein kinase pathway in a PDGF-dependent manner. Of note, TNIIIA2 induced continuous proliferation in NIH3T3 in an integrin α5ß1-dependent manner even after they formed a confluent monolayer. Thus, it was proposed that tenascin-C might be involved in deregulated cell growth through potentiated and sustained activation of integrin α5ß1 after exposure of the proadhesive effect of TNIIIA2.


Assuntos
Proliferação de Células/efeitos dos fármacos , Peptídeos/farmacologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Vitronectina/metabolismo , Tenascina/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Humanos , Células K562 , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Camundongos , Células NIH 3T3 , Peptídeos/química , Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Vitronectina/genética , Sindecana-4/genética , Sindecana-4/metabolismo , Tenascina/química
11.
Apoptosis ; 20(6): 843-57, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25690319

RESUMO

As a glycol-protein located in extracellular matrix (ECM), tenascin-C (TNC) is absent in most normal adult tissues but is highly expressed in the majority of malignant solid tumors. Pancreatic cancer is characterized by an abundant fibrous tissue rich in TNC. Although it was reported that TNC's expression increased in the progression from low-grade precursor lesions to invasive cancer and was associated with tumor differentiation in human pancreatic cancer, studies on the relations between TNC and tumor progression in pancreatic cancer were rare. In this study, we performed an analysis to determine the effects of TNC on modulating cell apoptosis and chemo-resistance and explored its mechanisms involving activation in pancreatic cancer cell. The expressions of TNC, ERK1/2/p-ERK1/2, Bcl-xL and Bcl-2 were detected by immunohistochemistry and western blotting. Then the effects of exogenous and endogenous TNC on the regulation of tumor proliferation, apoptosis and gemcitabine cytotoxicity were investigated. The associations among the TNC knockdown, TNC stimulation and expressions of ERK1/2/NF-κB/p65 and apoptotic regulatory proteins were also analyzed in cell lines. The mechanism of TNC on modulating cancer cell apoptosis and drug resistant through activation of ERK1/2/NF-κB/p65 signals was evaluated. The effect of TNC on regulating cell cycle distribution was also tested. TNC, ERK1/2/p-ERK1/2, and apoptotic regulatory proteins Bcl-xL and Bcl-2 were highly expressed in human pancreatic cancer tissues. In vitro, exogenous TNC promoted pancreatic cancer cell growth also mediates basal as well as starved and drug-induced apoptosis in pancreatic cancer cells. The effects of TNC on anti-apoptosis were induced by the activation state of ERK1/2/NF-κB/p65 signals in pancreatic cell. TNC phosphorylate ERK1/2 to induce NF-κB/p65 nucleus translocation. The latter contributes to promote Bcl-xL, Bcl-2 protein expressions and reduce caspase activity, which inhibit cell apoptotic processes. TNC mediated gemcitabine chemo-resistance via modulating cell apoptosis in pancreatic cancer. TNC resulted in the enrichment of pancreatic cancer cells in S-phase with a concomitant decrease in number of cells in G1 phase. The present study indicated TNC in cellular matrix induces an activation of ERK1/2/NF-κB/p65 signaling cascade and thereby mediates resistance to apoptosis in pancreatic cancer. TNC could serve as a diagnostic marker and predictor of gemcitabine response and potentially as a target for chemotherapy of pancreatic cancer.


Assuntos
Adenocarcinoma/metabolismo , Apoptose/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias Pancreáticas/metabolismo , Tenascina/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistência a Medicamentos/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo
12.
Acta Neurochir Suppl ; 120: 99-103, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25366607

RESUMO

BACKGROUND AND PURPOSE: The authors have reported that tenascin-C (TNC), a matricellular protein, is induced after subarachnoid hemorrhage (SAH), associated with cerebral vasospasm. In this study, we examined whether TNC alone causes cerebral vasospasm-like constriction of the intracranial internal carotid arteries (ICAs) in rats, focusing on the p38 mitogen-activated protein kinase (MAPK)-mediated mechanisms. METHODS: First, we injected 10 µg of TNC into the cisterna magna of healthy rats and studied morphologically whether TNC caused constriction of the left ICA at 24-72 h after administration. Second, we examined the effect of SB203580 (a p38 MAPK inhibitor) on the vessel diameter of the left ICA in healthy rats at 24 h. Third, we evaluated the effect of SB203580 on TNC-induced constriction of the left ICA in healthy rats at 24 h. RESULTS: TNC significantly induced cerebral vasospasm-like angiographic constriction of the left ICAs, which continued at least for 72 h. SB203580 itself had no effect on the diameter of normal ICAs, but abolished the TNC-induced vasoconstrictive effect on the left ICA. CONCLUSION: These findings show that TNC causes left ICA constriction via activation of p38 MAPK, resembling post-SAH vasospasm, and suggest the possible involvement of TNC in the pathogenesis of cerebral vasospasm.


Assuntos
Artéria Carótida Interna/efeitos dos fármacos , Hemorragia Subaracnóidea/tratamento farmacológico , Tenascina/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoespasmo Intracraniano/tratamento farmacológico , Animais , Artéria Carótida Interna/fisiologia , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Masculino , Piridinas/farmacologia , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/fisiopatologia , Tenascina/fisiologia , Vasoconstrição/fisiologia , Vasoespasmo Intracraniano/fisiopatologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Rheumatology (Oxford) ; 53(3): 415-24, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24241034

RESUMO

OBJECTIVE: The role of the joint tissue microenvironment in the pathogenesis of human RA has recently attracted much attention. The present study investigated the roles of α9ß1 integrin and its ligands in synovial specimens of human RA patients in generating the unique human arthritic tissue microenvironment. METHODS: Synovial fibroblasts and macrophages were isolated from the synovial tissue of patients with RA or OA. The expression of α9ß1 integrin was analysed using FACS with multicolour staining. The production of MMPs and proinflammatory cytokines was analysed in cultures of synovial fibroblasts and macrophages with α9ß1 integrin ligands. RESULTS: Synovial fibroblasts and macrophages derived from arthritic joints spontaneously secreted tenascin-C and osteopontin. Synovial fibroblasts and macrophages obtained from patients with RA expressed α9ß1 integrins, a common receptor for osteopontin and tenascin-C. In the synovial fibroblasts of RA, the amount of tenascin-C protein produced was much greater than that of osteopontin in synovial fibroblasts of RA. Importantly, autocrine and paracrine interactions of α9ß1 integrin and tenascin-C induced the expression of MMPs and IL-6 in synovial fibroblasts, as well as TNF-α and IL-1ß in synovial macrophages. CONCLUSION: These findings indicate that autocrine and paracrine interaction of α9ß1 integrin and tenascin-C in the joint tissue microenvironment contributes to the pathogenesis of RA. Therefore α9ß1 integrin may become a potential therapeutic target for RA.


Assuntos
Artrite Reumatoide/patologia , Artrite Reumatoide/fisiopatologia , Integrinas/fisiologia , Osteoartrite/patologia , Osteoartrite/fisiopatologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Tenascina/farmacologia , Regulação para Cima/genética
14.
Biomacromolecules ; 15(12): 4480-7, 2014 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-25343209

RESUMO

Extracellular matrix contains various signals for cell surface receptors that regulate cell fate through modulation of cellular activities such as proliferation and differentiation. Cues from extracellular matrix components can be used for development of new materials to control the stem cell fate. In this study, we achieved control of stem cell fate toward osteogenic commitment by using a single extracellular matrix element despite the contradictory effect of mechanical stiffness. For this purpose, we mimicked bone extracellular matrix by incorporating functional sequence of fibronectin type III domain from native tenascin-C on self-assembled peptide nanofibers. When rat mesenchymal stem cells (rMSCs) were cultured on these peptide nanofibers, alkaline phosphatase (ALP) activity and alizarin red staining indicated osteogenic differentiation even in the absence of osteogenic supplements. Moreover, expression levels of osteogenic marker genes were significantly enhanced revealed by quantitative real-time polymerase chain reaction (qRT-PCR), which showed the remarkable bioactive role of this nanofiber system on osteogenic differentiation. Overall, these results showed that tenascin-C mimetic peptides significantly enhanced the attachment, proliferation, and osteogenic differentiation of rMSCs even in the absence of any external bioactive factors and regardless of the suitable stiff mechanical properties normally required for osteogenic differentiation. Thus, these peptide nanofibers provide a promising new platform for bone regeneration.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Nanofibras/química , Osteogênese/efeitos dos fármacos , Tenascina/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Regeneração Óssea/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Matriz Extracelular/metabolismo , Expressão Gênica , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão e Varredura , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reologia
15.
Acta Neurochir Suppl ; 115: 213-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22890671

RESUMO

INTRODUCTION: Matricellular protein (MCP) is a class of nonstructural and secreted extracellular matrix proteins that exert diverse functions, but its role in vascular smooth muscle contraction has not been investigated. MATERIAL AND METHODS: First, rat subarachnoid hemorrhage (SAH) models were produced by endovascular perforation and examined for tenascin-C (TNC) and osteopontin (OPN) induction (representatives of MCPs) in vasospastic cerebral arteries using immunostaining. Second, recombinant TNC (r-TNC), recombinant OPN (r-OPN), or both were injected into a cisterna magna in healthy rats, and the effects on the diameter of basilar arteries were determined using India ink angiography. RESULTS: In SAH rats, TNC immunoreactivity was markedly induced in the smooth muscle cell layers of spastic cerebral arteries on day 1 but not in control animals. The TNC immunoreactivity decreased on day 3 as vasospasm improved: OPN immunoreactivity, on the other hand, was more induced in the arterial wall on day 3. r-TNC injections caused prolonged contractions of rat basilar arteries, which were reversed by r-OPN, although r-OPN itself had no effect on the vessel diameter. CONCLUSIONS: MCPs, including TNC and OPN, may contribute to the pathophysiology of cerebral vasospasm and provide a novel therapeutic approach against it.


Assuntos
Osteopontina/metabolismo , Hemorragia Subaracnóidea/complicações , Tenascina/metabolismo , Vasoespasmo Intracraniano/etiologia , Animais , Artéria Basilar/efeitos dos fármacos , Artéria Basilar/metabolismo , Artéria Basilar/patologia , Carbono , Angiografia Cerebral , Cisterna Magna/efeitos dos fármacos , Cisterna Magna/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Exame Neurológico , Osteopontina/farmacologia , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Hemorragia Subaracnóidea/etiologia , Tenascina/farmacologia , Fatores de Tempo , Vasoconstrição/efeitos dos fármacos , Vasoespasmo Intracraniano/patologia
16.
Int Endod J ; 46(1): 30-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22747576

RESUMO

AIM: To investigate the effects of tenascin-C (TN-C) on cultured rat dental pulp cells in relation to the expression of Notch signalling. METHODOLOGY: Subcultured dental pulp cells derived from rat incisors were seeded both in wells and on plastic coverslips coated with various concentrations of recombinant human TN-C. Expression of bone-related mRNA was then analysed by RT-PCR and observed by immunohistochemical staining. Encoding of Notch1 and Notch2 (markers of initial differentiation of odontoblast-like cells), alkaline phosphatase (ALP), osteopontin (OPN) and osteocalcin (OCN) (markers of mineralization) was investigated. Non-TN-C-coated wells were used as controls. Primary antibodies to Notch1, ALP and OCN were used for immunofluorescence staining, and ALP activity was evaluated. Data were compared using Student's t-test. RESULTS: Cell proliferation rate in the experimental groups was significantly higher (P < 0.05) than that in the control group at 72 h. Expression of Notch1, Notch2, ALP, OPN and OCN mRNAs was significantly higher (P < 0.05) in the experimental group than that in the control group. Strongly positive staining for Notch1, ALP and OCN was observed in the experimental group. ALP activity was significantly higher (P < 0.01) in the experimental group than in the control group at 24 h. CONCLUSION: TN-C promoted differentiation of rat dental pulp cells by the activation of Notch.


Assuntos
Polpa Dentária/efeitos dos fármacos , Tenascina/farmacologia , Fosfatase Alcalina/análise , Animais , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular , Células Cultivadas , Polpa Dentária/citologia , Imunofluorescência , Humanos , Masculino , Odontoblastos/efeitos dos fármacos , Osteocalcina/análise , Osteopontina/análise , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptor Notch1/análise , Receptor Notch2/análise , Proteínas Recombinantes , Fatores de Tempo
17.
J Plast Reconstr Aesthet Surg ; 83: 69-76, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37270997

RESUMO

BACKGROUND: Although autologous fat grafting is considered a successful method for the management of contour deformities, the fat graft could potentially induce cancer reappearance by fueling dormant breast cancer cells. Our aim was to characterize the role of adipose-derived stem cells on active and dormant breast cancer cell growth. METHODS: Cobalt chloride was used to induce dormancy in MCF-7 cancer cells. Proliferation of active and dormant cancer cells was determined in the presence of adipose-derived stem cells. A proteome array was used to detect cancer-related protein expression in the cell-conditioned medium. The migration of cancer cells was measured in response to conditioned medium from the adipose-derived stem cells. RESULTS: The adipose-derived stem cells showed variable effects on active MCF-7 cells growth and inhibited MCF-7 proliferation after the withdrawal of cobalt chloride. Of the 84 different proteins measured in the conditioned medium, only tenascin-C was differentially expressed in the co-cultures. MCF-7 cells alone did not express tenascin-C, whereas co-cultures between MCF-7 and adipose-derived stem cells expressed more tenascin-C versus adipose-derived stem cells alone. The conditioned medium from co-cultures significantly increased the migration of the cancer cells. CONCLUSIONS: Adipose-derived stem cells themselves neither increased the growth or migration of cancer cells, suggesting that autologous fat grafting may be oncologically safe if reconstruction is postponed until there is no evidence of active disease. However, interactions between adipose-derived stem cells and MCF-7 cancer cells could potentially lead to the production of factors, which further promote cancer cell migration.


Assuntos
Tecido Adiposo , Neoplasias da Mama , Humanos , Feminino , Tecido Adiposo/transplante , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Tenascina/metabolismo , Tenascina/farmacologia , Células-Tronco , Proliferação de Células
18.
Adv Mater ; 35(33): e2301493, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37227134

RESUMO

The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that match the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro. Here, a synthetic, bioactive hydrogel is synthesized that mimics the native lung modulus and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin-binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which enables quiescent culture of human lung fibroblasts (HLFs). Stimulation with transforming growth factor ß1 (TGF-ß1), metastatic breast cancer conditioned media (CM), or tenascin-C-derived integrin-binding peptide activated hydrogel-encapsulated HLFs demonstrates multiple environmental methods to activate HLFs in a lung ECM-mimicking hydrogel. This lung hydrogel platform is a tunable, synthetic approach to studying the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.


Assuntos
Neoplasias da Mama , Tenascina , Humanos , Feminino , Tenascina/metabolismo , Tenascina/farmacologia , Matriz Extracelular/metabolismo , Fibroblastos , Hidrogéis/química , Peptídeos/química , Pulmão , Integrinas/metabolismo
19.
Neurobiol Dis ; 46(1): 172-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22300707

RESUMO

Platelet-derived growth factor (PDGF) has been implicated in the pathogenesis of cerebral vasospasm after subarachnoid hemorrhage (SAH), but the mechanism remains unknown. The purpose of this study was to assess whether imatinib mesylate (imatinib), an inhibitor of the tyrosine kinases of PDGF receptors (PDGFRs), prevents cerebral vasospasm after SAH in rats, and to elucidate if tenascin-C (TNC), a matricellular protein, is involved in the mechanism. Imatinib (10 or 50 mg/kg body weight) was administered intraperitoneally to rats undergoing SAH by endovascular perforation, and the effects were evaluated by neurobehavioral tests and India-ink angiography at 24-72 h post-SAH. Western blotting and immunohistochemistry were performed to explore the underlying mechanisms in cerebral arteries at 24h post-SAH. Recombinant TNC was administered intracisternally to imatinib-treated SAH rats, and the effects were evaluated by neurobehavioral tests, India-ink angiography and immunohistochemistry at 24 h post-SAH. Both dosages of imatinib significantly prevented post-SAH neurological impairments and vasospasm at 24-72 h. SAH caused PDGFR-ß upregulation, PDGFR activation, mitogen-activated protein kinase activation, and TNC upregulation in the spastic cerebral arteries, all of which were significantly suppressed by imatinib treatment. Recombinant TNC reversed the anti-vasospastic effects and protein expression changes by imatinib. This study suggests that imatinib prevents cerebral vasospasm at least partly via inhibiting the upregulation of TNC, implying that TNC may be a new therapeutic target for post-SAH vasospasm.


Assuntos
Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Hemorragia Subaracnóidea/tratamento farmacológico , Tenascina/metabolismo , Vasoespasmo Intracraniano/prevenção & controle , Animais , Benzamidas , Modelos Animais de Doenças , Mesilato de Imatinib , Masculino , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/patologia , Tenascina/farmacologia , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/metabolismo
20.
Am J Pathol ; 178(2): 754-63, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21281808

RESUMO

Tenascin C (TNC) is an extracellular matrix glycoprotein up-regulated in solid tumors. Higher TNC expression is shown in invading fronts of breast cancer, which correlates with poorer patient outcome. We examined whether TNC induces epithelial-mesenchymal transition (EMT) in breast cancer. Immunohistochemical analysis of invasive ductal carcinomas showed that TNC deposition was frequent in stroma with scattered cancer cells in peripheral margins of tumors. The addition of TNC to the medium of the MCF-7 breast cancer cells caused EMT-like change and delocalization of E-cadherin and ß-catenin from cell-cell contact. Although amounts of E-cadherin and ß-catenin were not changed after EMT in total lysates, they were increased in the Triton X-100-soluble fractions, indicating movement from the membrane into the cytosol. In wound healing assay, cells were scattered from wound edges and showed faster migration after TNC treatment. The EMT phenotype was correlated with SRC activation through phosphorylation at Y418 and phosphorylation of focal adhesion kinase (FAK) at Y861 and Y925 of SRC substrate sites. These phosphorylated proteins colocalized with αv integrin-positive adhesion plaques. A neutralizing antibody against αv or a SRC kinase inhibitor blocked EMT. TNC could induce EMT-like change showing loss of intercellular adhesion and enhanced migration in breast cancer cells, associated with FAK phosphorylation by SRC; this may be responsible for the observed promotion of TNC in breast cancer invasion.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Tenascina/farmacologia , Quinases da Família src/metabolismo , Anticorpos Neutralizantes/farmacologia , Caderinas/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Feminino , Humanos , Integrina alfaV/imunologia , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Frações Subcelulares/metabolismo , Cicatrização/efeitos dos fármacos , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA