Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
1.
Invest Ophthalmol Vis Sci ; 63(1): 27, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-35060996

RESUMO

Purpose: Erianin has been reported to inhibit tumor activity by suppressing the expression of integrins. It is hypothesized that erianin can inhibit retinal neovascularization in collagen by suppressing the expression of integrins. With an aim to test this hypothesis, the regulation of erianin on collagen-mediated retinal angiogenesis via the Ras homolog gene family member A (RhoA)/Rho-associated coiled-coil containing protein kinase 1 (ROCK1) signaling pathway induced by α2 and ß1 integrin-collagen interactions was investigated. Methods: The effects of erianin on human retinal vascular endothelial cells (HRVECs) were assessed in vitro using a hypoxia model in a three-dimensional cell culture induced by cobalt (II) chloride (CoCl2). A hypoxia-induced retinopathy model in adult zebrafish and zebrafish embryos was established to assess the antiangiogenic effect of erianin with and without vitreous collagen in vivo. The expression of α2 and ß1 integrin and RhoA/ROCK1 pathway in HRVECs and zebrafish retinas were analyzed. Results: In vitro, collagen improved the angiogenic potential of HRVECs, including migration, adhesion, and tube formation, in a three-dimensional cell culture model. Erianin suppressed the angiogenic processes of the CoCl2-induced hypoxia HRVEC model in a concentration-dependent manner. In vivo, erianin reduced retinal angiogenesis in the hypoxia-induced retinopathy model in adult and embryo zebrafish. Erianin inhibited the expression of α2 and ß1 integrin and RhoA/ROCK1 in a hypoxia-induced model in vitro in three-dimensional cell culture and in vivo in adult zebrafish. Conclusions: Collagen-mediated retinal angiogenesis may be regulated by erianin via the RhoA/ROCK1 signaling pathway induced by α2 and ß1 integrin-collagen interactions. These findings suggest that erianin has the therapeutic potential on intraocular collagen-mediated retinal angiogenesis.


Assuntos
Bibenzilas/farmacologia , Regulação da Expressão Gênica , Integrina alfa1/genética , Integrina beta1/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Fenol/farmacologia , Neovascularização Retiniana/genética , Proteínas de Peixe-Zebra/genética , Quinases Associadas a rho/genética , Animais , Movimento Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Integrina alfa1/biossíntese , Integrina beta1/biossíntese , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese , Quinases Associadas a rho/biossíntese
2.
J Biomed Mater Res B Appl Biomater ; 109(11): 1857-1865, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33847460

RESUMO

Tenon's capsule fibroblasts are the main cellular components of filtration tract scar that limit the success rate of glaucoma filtration surgery. Scar formation results from infiltration and proliferation of fibroblasts into damaged areas, meanwhile synthesis of extracellular matrix glycoproteins. Integrins are cell surface receptors for extracellular molecules that mediate cell adhesion, spreading, migration, and invasion. They bind their ligands often through recognition of short amino-acid sequences-arginine-glycine-aspartic acid (RGD). Peptides that contain RGD sequence can compete with RGD containing insoluble matrix proteins for binding to the integrin receptor and thus prevent the downstream signaling pathway. Increasing evidence supports that ß1-integrin/focal adhesion kinase (FAK)/Akt signal pathway plays an important role in fibrogenesis and scar formation in different tissues. In consideration of advantages of peptide hydrogel, that is well biocompatibility, gel state, degradability, good drug loading, we designed and fabricated an RGD peptide hydrogel, and hypothesized that it could inhibit the expression of ß1-integrin, FAK, and Akt in Tenon's capsule fibroblasts. Rheology results showed that 1% wt Fmoc-FFGGRGD peptide solution could self-assemble into hydrogel. Western blot analysis revealed that there were statistical differences between control group and 1% wt group in ß1-integrin/ß-actin, FAK/ß-actin, Akt/ß-actin respectively (*p < .05). The relative mRNA expression of ß1-integrin, FAK, Akt in control group and 1% wt group were also statistically different respectively (*p < .05). We proved that 1% wt Fmoc-FFGGRGD self-assembly peptide hydrogel could inhibit the expression of ß1-integrin, FAK and Akt in Tenon's capsule fibroblasts. It is a promising way to solve scar formation of glaucoma filter channel.


Assuntos
Fibroblastos/metabolismo , Quinase 1 de Adesão Focal/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Hidrogéis , Integrina beta1/biossíntese , Oligopeptídeos , Proteínas Proto-Oncogênicas c-akt/biossíntese , Tendões/metabolismo , Animais , Hidrogéis/química , Hidrogéis/farmacologia , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley
3.
J Cell Mol Med ; 24(18): 10792-10802, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32803867

RESUMO

Brain-derived neurotrophic factor (BDNF) has been reported to participate in fracture healing, whereas the mechanism is still unclear. Since osteoblast migration is important for fracture healing, investigating effects of BDNF on osteoblasts migration may help to reveal its mechanism. Here, MC3T3-E1 cells were used in vitro while closed femur fracture mice were applied in vivo. Cells migration was assessed with Transwell assay. The protein expression was analysed by immunoblotting. X-ray and Micro-CT were performed at different time after fracture. Our results showed that BDNF promoted MC3T3-E1 cells migration, integrin ß1 expression and ERK1/2 and AKT phosphorylation. K252a, a specific inhibitor for TrkB, suppressed BDNF-induced migration, integrin ß1 expression and activation of ERK1/2 and AKT. PD98059 (an ERK1/2 inhibitor) and LY294002 (an AKT inhibitor) both inhibited BDNF-induced migration and integrin ß1 expression while integrin ß1 blocking antibody only suppressed cell migration. X-ray and Micro-CT analyses showed that the adenoviral carried integrin ß1 shRNA group had slower fracture healing at 7 and 21 days, but not 35 days compared to the control group. Thus, we proposed that BDNF stimulated MC3T3-E1 cells migration by up-regulating integrin ß1 via TrkB mediated ERK1/2 and AKT signalling, and this may help to enhance the fracture healing.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Consolidação da Fratura/efeitos dos fármacos , Integrina beta1/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Glicoproteínas de Membrana/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Linhagem Celular Transformada , Movimento Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Fraturas do Fêmur/diagnóstico por imagem , Fraturas do Fêmur/tratamento farmacológico , Fraturas do Fêmur/fisiopatologia , Integrina beta1/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/química , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Regulação para Cima/efeitos dos fármacos , Microtomografia por Raio-X
4.
Stem Cells ; 37(12): 1528-1541, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31574184

RESUMO

Nearly 1 in every 120 children born has a congenital heart defect. Although surgical therapy has improved survival, many of these children go on to develop right ventricular heart failure (RVHF). The emergence of cardiovascular regenerative medicine as a potential therapeutic strategy for pediatric HF has provided new avenues for treatment with a focus on repairing or regenerating the diseased myocardium to restore cardiac function. Although primarily tried using adult cells and adult disease models, stem cell therapy is relatively untested in the pediatric population. Here, we investigate the ability of electrical stimulation (ES) to enhance the retention and therapeutic function of pediatric cardiac-derived c-kit+ progenitor cells (CPCs) in an animal model of RVHF. Human CPCs isolated from pediatric patients were exposed to chronic ES and implanted into the RV myocardium of rats. Cardiac function and cellular retention analysis showed electrically stimulated CPCs (ES-CPCs) were retained in the heart at a significantly higher level and longer time than control CPCs and also significantly improved right ventricular functional parameters. ES also induced upregulation of extracellular matrix and adhesion genes and increased in vitro survival and adhesion of cells. Specifically, upregulation of ß1 and ß5 integrins contributed to the increased retention of ES-CPCs. Lastly, we show that ES induces CPCs to release higher levels of pro-reparative factors in vitro. These findings suggest that ES can be used to increase the retention, survival, and therapeutic effect of human c-kit+ progenitor cells and can have implications on a variety of cell-based therapies. Stem Cells 2019;37:1528-1541.


Assuntos
Estimulação Elétrica/métodos , Insuficiência Cardíaca/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/métodos , Função Ventricular Direita/fisiologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Pré-Escolar , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Cardiopatias Congênitas/cirurgia , Humanos , Lactente , Recém-Nascido , Integrina beta1/biossíntese , Masculino , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Medicina Regenerativa/métodos , Células-Tronco/citologia
5.
Biochem Biophys Res Commun ; 514(4): 1115-1121, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31101337

RESUMO

Cancer cells can invade as a population in various cancer tissues. This phenomenon is called collective invasion, which is associated with the metastatic potential and prognosis of cancer patients. The collectiveness of cancer cells is necessary for collective invasion. However, the mechanism underlying the generation of collectiveness by cancer cells is not well known. In this study, the phenomenon of contact following, where neighboring cells move in the same direction via intercellular adhesion, was investigated. An experimental system was created to observe the two-dimensional invasion using a collagen gel overlay to study contact following in collective invasion. The role of integrin-ß1, one of the major extracellular matrix (ECM) receptors, in contact following was examined through the experimental system. Integrin-ß1 was localized to the intercellular site in squamous carcinoma cells. Moreover, the intercellular adhesion and contact following were suppressed by treatment of an integrin-ß1 inhibitory antibody. ECM proteins such as laminin-332 and type-XVII collagen were also localized to the intercellular site and critical for contact following. Collectively, it was demonstrated that the activity of integrin-ß1 and expression of ECM proteins in the intercellular site promote contact following in the collective invasion of a cancer cell population.


Assuntos
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adesão Celular/metabolismo , Integrina beta1/metabolismo , Colágenos não Fibrilares/metabolismo , Neoplasias Cutâneas/metabolismo , Autoantígenos/biossíntese , Carcinoma de Células Escamosas/patologia , Adesão Celular , Moléculas de Adesão Celular/biossíntese , Humanos , Integrina beta1/biossíntese , Colágenos não Fibrilares/biossíntese , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas , Calinina , Colágeno Tipo XVII
6.
Int J Cancer ; 145(3): 830-841, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30719702

RESUMO

Breast cancer remains the second cause of tumor-related mortality in women worldwide mainly due to chemoresistance and metastasis. The chemoresistance and metastasis are attributed to a rare subpopulation with enriched stem-like characteristics, thus called Cancer Stem Cells (CSCs). We have previously reported aberrant expression of the actin-bundling protein (fascin) in breast cancer cells, which enhances their chemoresistance, metastasis and enriches CSC population. The intracellular mechanisms that link fascin with its downstream effectors are not fully elucidated. Here, loss and gain of function approaches in two different breast cancer models were used to understand how fascin promotes disease progression. Importantly, findings were aligned with expression data from actual breast cancer patients. Expression profiling of a large breast cancer dataset (TCGA, 530 patients) showed statistically significant correlation between fascin expression and a key adherence molecule, ß1 integrin (ITGB1). In vitro manipulation of fascin expression in breast cancer cells exhibited its direct effect on ITGB1 expression. Fascin-mediated regulation of ITGB1 was critical for several breast cancer cell functions including adhesion to different extracellular matrix, self-renewability and chemoresistance. Importantly, there was a significant relationship between fascin and ITGB1 co-expression and short disease-free as well as overall survival in chemo-treated breast cancer patients. This novel role of fascin effect on ITGB1 expression and its outcome on cell self-renewability and chemoresistance strongly encourages for dual targeting of fascin-ITGB1 axis as a therapeutic approach to halt breast cancer progression and eradicate it from the root.


Assuntos
Neoplasias da Mama/patologia , Proteínas de Transporte/biossíntese , Integrina beta1/biossíntese , Proteínas dos Microfilamentos/biossíntese , Células-Tronco Neoplásicas/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Integrina beta1/genética , Integrina beta1/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Análise de Sobrevida , Regulação para Cima
7.
J Biomed Mater Res A ; 107(3): 663-677, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30474276

RESUMO

In biomaterials research, biomechanics which support tissue regeneration steadily gains of importance. Hence, we have previously shown that gelatin-based electrospun nonwoven mats (NWMs) with a distinct modulus of elasticity (3.2 kPa) promotes epithelial morphogenesis. Since molecular mechanisms of this morphogenesis are still unknown, the present study aims at identifying molecules, involved herein. Epithelia established on the NMWs showed persistence of the activated state of the epidermal growth factor receptor (EGF-R), phosphorylated at the src-specific tyrosine 845 (EGF-RT845 ) throughout the observation period of 10 days. To elucidate whether the observed morphogenesis mechanistically involves EGF-R signaling, we inhibited EGF-R, by employing the EGF-RT845 specific inhibitor Gefitinib (IRESSA®). Gefitinib administration yielded a reduced expression of the ß1 integrin subunit, a well-known cell-matrix interaction receptor, concomitant with downregulation of p42/44 ERK1/2 MAP-kinase activity. To elucidate whether the observed downregulation of ß1 is EGF-RT845 -dependent or emerging from ERK1/2 signaling, we exposed epithelia, grown on the NWMs, with the ERK1/2-directed inhibitor U0126. In the absence of Gefitinib, inhibition of p42/44 MAP-kinase activity resulted in decreased ß1 integrin protein levels, thus indicating that ß1 expression is dependent on ERK1/2 and not EGF-RT845 . Our results showed the first time that an EGF-R-ß1 integrin-signaling axis, including ERK1/2, promotes NWM-elasticity-based epithelial morphogenesis. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 663-677, 2019.


Assuntos
Fibroblastos/metabolismo , Gelatina , Queratinócitos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Butadienos/farmacologia , Linhagem Celular Transformada , Epitélio/metabolismo , Receptores ErbB/metabolismo , Fibroblastos/citologia , Gefitinibe/farmacologia , Gelatina/química , Gelatina/farmacologia , Humanos , Integrina beta1/biossíntese , Queratinócitos/citologia , Nitrilas/farmacologia
8.
Mater Sci Eng C Mater Biol Appl ; 93: 437-444, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30274076

RESUMO

Titanium (>99.6% purity) and its anodically oxidized modifications, with and without deposited silver nanodendrites regarding its biocompatibility were evaluated. In human gingival fibroblasts and osteoblast cell lines grown on tested samples, the level of expression of genes encoding αV (ITGAV) and ß1 (ITGB1) integrin subunits also genes encoding focal adhesion (FAK) and extracellular-signal regulated (ERK) kinases was assessed. For this purpose, the qualitative and quantitative PCR technique was used. The expression of studied genes was dependent on the origin of cell lines and the type of evaluated material. The high expression of PBGD and ITGAV genes in fibroblasts grown on the surface of anodically modified titanium with deposited silver nanodendrites indicates potentially high biocompatibility of these samples for soft tissue cells. The high expression of the ITGB1 and ERK1 genes and the enhanced expression of the FAK gene in osteoblasts cells grown on the tested material was also observed. Summarizing, the nanocrystalline Ti modified with silver deposits showed higher biocompatibility in comparison with the conventional pure Ti samples.


Assuntos
Materiais Revestidos Biocompatíveis , Dendrímeros , Fibroblastos/metabolismo , Teste de Materiais , Nanopartículas Metálicas/química , Osteoblastos/metabolismo , Prata , Titânio , Linhagem Celular Tumoral , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Dendrímeros/química , Dendrímeros/farmacologia , Fibroblastos/citologia , Quinase 1 de Adesão Focal/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Integrina alfa5/biossíntese , Integrina beta1/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Osteoblastos/citologia , Prata/química , Prata/farmacologia , Titânio/química , Titânio/farmacologia
9.
Exp Cell Res ; 353(2): 88-99, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28284838

RESUMO

In a previous transcriptomic analysis of 63 ocular melanomas of the uvea, we found that expression of the PRL-3/PTP4A3 gene, encoding a phosphatase that is anchored to the plasma membrane, was associated with the risk of metastasis, and a poor prognosis. We also showed that PRL-3 overexpression in OCM-1 ocular melanoma cells significantly increased cell migration in vitro and invasiveness in vivo, suggesting a direct role for PRL-3 in the metastatic spreading of uveal melanoma. Here, we aimed to identify PRL-3 substrates at the plasma membrane involved in adhesion to the extracellular matrix. We focused on integrin ß1, which is the most highly expressed integrin in our cohort of uveal melanomas. We show that preventing PRL-3 anchorage to the plasma membrane i) abolishes PRL-3-induced migration in OCM-1 cells, ii) specifically enhances the spreading of OCM-1 cells overexpressing PRL-3, and iii) favors the maturation of large focal adhesions (FAs) containing integrin ß1 on collagen I. Knockdown experiments confirmed integrin ß1 involvement in PRL3-induced migration. We identified interactions between PRL-3 and integrin ß1, as well as with FAK P-Y397, an auto-activated form of Focal Adhesion Kinase found in FAs. We also show that integrin ß1 may be dephosphorylated by PRL-3 in its intracytoplasmic S/T region, an important motif for integrin-mediated cell adhesion. Finally, we observed that PRL-3 regulated the clustering of integrin ß1 in FAs on collagen I but not on fibronectin. This work identifies PRL-3 as a new regulator of cell adhesion structures to the extracellular matrix, and further supports PRL-3 as a key actor of metastasis in uveal melanoma, of which molecular mechanisms are still poorly understood.


Assuntos
Movimento Celular/genética , Integrina beta1/biossíntese , Melanoma/genética , Proteínas de Neoplasias/biossíntese , Proteínas Tirosina Fosfatases/biossíntese , Neoplasias Uveais/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/patologia , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Integrina beta1/genética , Melanoma/patologia , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatases/genética , Neoplasias Uveais/patologia
10.
J Cell Biochem ; 118(9): 2693-2699, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28112417

RESUMO

Breast cancer is a complex and multifactorial disease. Tumors have a heterogeneous microenvironment, which have multiple interactions with other cell types, greatly influencing the behavior of tumor cells and response to therapy. The 3D culture mimics the microenvironment better found in vivo and is more appropriated than the traditional 2D culture made from plastic to test the cellular response to drugs. To investigate the effects of [10]-gingerol on breast tumor cells, we used physiologically relevant three-dimensional (3D) cultures of malignant and non-malignant human breast cells grown in laminin-rich extracellular matrix gels (lr-ECM). Our results showed selective cytotoxicity of [10]-gingerol against the malignant T4-2 breast cancer cell line compared to non-malignant S1 cells. The compound reverted the malignant phenotype of the cancer cells, downregulating the expression of epidermal growth factor receptor (EGFR) and ß1-integrin. Moreover, [10]-gingerol induced apoptosis in this cell line. These results suggest that [10]-gingerol may be an effective compound to use as adjuvant therapy in breast cancer treatment. J. Cell. Biochem. 118: 2693-2699, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Álcoois Graxos/farmacologia , Guaiacol/análogos & derivados , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Receptores ErbB/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Guaiacol/farmacologia , Humanos , Integrina beta1/biossíntese , Camundongos Nus , Proteínas de Neoplasias/biossíntese
11.
Clin Exp Metastasis ; 33(8): 839-852, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27612856

RESUMO

Epithelial ovarian cancer (EOC) dissemination is primarily mediated by the shedding of tumor cells from the primary site into ascites where they form multicellular spheroids that rapidly lead to peritoneal carcinomatosis. While the clinical importance and fundamental role of multicellular spheroids in EOC is increasingly appreciated, the mechanisms that regulate their formation and dictate their cellular composition remain poorly characterized. To investigate these important questions, we characterized spheroids isolated from ascites of women with EOC. We found that in these spheroids, a core of mesothelial cells was encased in a shell of tumor cells. Analysis further revealed that EOC spheroids are dynamic structures of proliferating, non-proliferating and hypoxic regions. To recapitulate these in vivo findings, we developed a three-dimensional co-culture model of primary EOC and mesothelial cells. Our analysis indicated that, compared to the OVCAR3 cell line, primary EOC cells isolated from ascites as well as mesothelial cells formed compact spheroids. Analysis of heterotypic spheroid microarchitecture revealed a structure that grossly resembles the structure of spheroids isolated from ascites. Cells that formed compact spheroids had elevated expression of ß1 integrin and low expression of E-cadherin. Addition of ß1 integrin blocking antibody or siRNA-mediated downregulation of ß1 integrin resulted in reduced tightness of the spheroids. Interestingly, the loss of MUC16 and E-cadherin expression resulted in the formation of more compact spheroids. Therefore, our findings support the heterotypic nature of spheroids from malignant EOC ascites. In addition, our data describe an unusual link between E-cadherin expression and less compact spheroids. Our data also emphasize the role of MUC16 and ß1 integrin in EOC spheroid formation.


Assuntos
Antígeno Ca-125/biossíntese , Caderinas/biossíntese , Integrina beta1/biossíntese , Proteínas de Membrana/biossíntese , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Neoplasias Peritoneais/patologia , Ascite/genética , Ascite/patologia , Líquido Ascítico/patologia , Antígeno Ca-125/genética , Caderinas/genética , Carcinoma/genética , Carcinoma/patologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Epitélio/metabolismo , Epitélio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina beta1/genética , Proteínas de Membrana/genética , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/secundário , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
12.
Oncol Rep ; 36(4): 2177-83, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27499367

RESUMO

An essential step in the peritoneal spread of ovarian cancer is the adhesion and implantation of tumor cells to the mesothelium layer. Integrin α5 and ß1 have been reported to mediate the initial adhesion process and to correlate with disease survival in ovarian cancer. However, the molecular mechanism of integrin α5ß1 dysregulation in tumorigenesis and metastasis remained enigmatic. In the present study, using the US NCI60 database, we identified miR-17 as a candidate regulator targeting both integrin α5 and ß1. The level of miR-17 was evidently inversely correlated with that of α5 and ß1 in ovarian cancer cell lines. Specifically, miR-17 bound directly to the 3' untranslated region (3'UTR) of α5 and ß1 and suppressed their expression. Forced expression of miR-17 led to markedly diminished adhesion and invasion of ovarian cancer cells in vitro, and notably reduced metastatic nodules inside the peritoneal cavity in in vivo SKOV3 xenografts model. Moreover, ectopic expression of miR-17 in ovarian cancer cells resulted in repressed ILK phosphorylation as well as decreased production of active matrix metalloproteinase-2 (MMP-2). Our results indicated that miR-17 hampered ovarian cancer peritoneal propagation by targeting integrin α5 and ß1. These findings supported the utility of miR-17/α5ß1 to be considered as valuable marker for metastatic potential of ovarian cancer cells, or a therapeutic target in ovarian cancer treatment.


Assuntos
Integrina alfaV/biossíntese , Integrina beta1/biossíntese , Metaloproteinase 2 da Matriz/biossíntese , MicroRNAs/genética , Neoplasias Ovarianas/genética , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina alfaV/genética , Integrina beta1/genética , Metaloproteinase 2 da Matriz/genética , Camundongos , MicroRNAs/biossíntese , Invasividade Neoplásica/genética , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cell Oncol (Dordr) ; 39(5): 449-462, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27376838

RESUMO

BACKGROUND: Previously, nitric oxide (NO) has been found to affect the metastatic behavior of various types of cancer. In addition, it has been found that alterations in integrin expression may have profound effects on cancer cell survival and migration. Here, we aimed at assessing the effects of non-toxic concentrations of NO on human non-small cell lung cancer (NSCLC) cells, including the expression of integrins and the migration of these cells. METHODS: The cytotoxic and proliferative effects of NO on human NSCLC-derived H460, H292 and H23 cells were tested by MTT assay. The migration capacities of these cells was evaluated by wound healing and transwell migration assays. The expression of integrins and migration-associated proteins was determined by Western blot analyses. RESULTS: We found that NO treatment caused a significant increase in the expression of integrin αv and ß1 in all three NSCLC-derived cell lines tested. Known migration-associated proteins acting downstream of these integrins, including focal adhesion kinase (FAK), active RhoA (Rho-GTP) and active cell division control 42 (Cdc42-GTP), were found to be significantly activated in response to NO. In addition, we found that NO-treated cells showed an increased motility and that this motility was associated with a significant increase in the number of filopodia per cell. We also found that NO-treated cells exhibited increased active protein kinase G (PKG), protein kinase B (AKT) and FAK expression levels. Using a pharmacological approach, we found that the integrin-modulating effect of NO is most likely brought about by a PKG/AKT-dependent mechanism, since the observed changes in integrin expression were abolished by AKT inhibitors, but not by FAK inhibitors. CONCLUSION: Our data suggest a novel role of NO in the regulation of integrin expression and, concomitantly, the migratory capacity of NSCLC cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Integrina alfaV/biossíntese , Integrina beta1/biossíntese , Neoplasias Pulmonares/patologia , Óxido Nítrico/farmacologia , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima
14.
Oncotarget ; 7(25): 38408-38426, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27224909

RESUMO

We have previously shown that stromal cells desensitize breast cancer cells to the anti-estrogen fulvestrant and, along with it, downregulate the expression of TMEM26 (transmembrane protein 26). In an effort to study the function and regulation of TMEM26 in breast cancer cells, we found that breast cancer cells express non-glycosylated and N-glycosylated isoforms of the TMEM26 protein and demonstrate that N-glycosylation is important for its retention at the plasma membrane. Fulvestrant induced significant changes in expression and in the N-glycosylation status of TMEM26. In primary breast cancer, TMEM26 protein expression was higher in ERα (estrogen receptor α)/PR (progesterone receptor)-positive cancers. These data suggest that ERα is a major regulator of TMEM26. Significant changes in TMEM26 expression and N-glycosylation were also found, when MCF-7 and T47D cells acquired fulvestrant resistance. Furthermore, patients who received aromatase inhibitor treatment tend to have a higher risk of recurrence when tumoral TMEM26 protein expression is low. In addition, TMEM26 negatively regulates the expression of integrin ß1, an important factor involved in endocrine resistance. Data obtained by spheroid formation assays confirmed that TMEM26 and integrin ß1 can have opposite effects in breast cancer cells. These data are consistent with the hypothesis that, in ERα-positive breast cancer, TMEM26 may function as a tumor suppressor by impeding the acquisition of endocrine resistance. In contrast, in ERα-negative breast cancer, particularly triple-negative cancer, high TMEM26 expression was found to be associated with a higher risk of recurrence. This implies that TMEM26 has different functions in ERα-positive and -negative breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proteínas de Membrana/biossíntese , Biomarcadores Farmacológicos/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/biossíntese , Feminino , Fulvestranto , Humanos , Integrina beta1/biossíntese , Células MCF-7 , Proteínas de Membrana/genética , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , RNA/genética , RNA/metabolismo
15.
Blood ; 128(1): 55-9, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207790

RESUMO

Krüppel-like factor 5 (Klf5) encodes a zinc-finger transcription factor and has been reported to be a direct target of C/EBPα, a master transcription factor critical for formation of granulocyte-macrophage progenitors (GMP) and leukemic GMP. Using an in vivo hematopoietic-specific gene ablation model, we demonstrate that loss of Klf5 function leads to a progressive increase in peripheral white blood cells, associated with increasing splenomegaly. Long-term hematopoietic stem cells (HSCs), short-term HSCs (ST-HSCs), and multipotent progenitors (MPPs) were all significantly reduced in Klf5(Δ/Δ) mice, and knockdown of KLF5 in human CD34(+) cells suppressed colony-forming potential. ST-HSCs, MPPs, and total numbers of committed progenitors were increased in the spleen of Klf5(Δ/Δ) mice, and reduced ß1- and ß2-integrin expression on hematopoietic progenitors suggests that increased splenic hematopoiesis results from increased stem and progenitor mobilization. Klf5(Δ/Δ) mice show a significant reduction in the fraction of Gr1(+)Mac1(+) cells (neutrophils) in peripheral blood and bone marrow and increased frequency of eosinophils in the peripheral blood, bone marrow, and lung. Thus, these studies demonstrate dual functions of Klf5 in regulating hematopoietic stem and progenitor proliferation and localization in the bone marrow, as well as lineage choice after GMP, promoting increased neutrophil output at the expense of eosinophil production.


Assuntos
Regulação da Expressão Gênica/fisiologia , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Multipotentes/metabolismo , Animais , Antígenos CD18/biossíntese , Antígenos CD18/genética , Eosinófilos/citologia , Eosinófilos/metabolismo , Células Progenitoras de Granulócitos e Macrófagos/citologia , Integrina beta1/biossíntese , Integrina beta1/genética , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Células-Tronco Multipotentes/citologia , Neutrófilos/citologia , Neutrófilos/metabolismo
16.
Cancer Sci ; 107(7): 916-23, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27094578

RESUMO

Increasing evidence has revealed that miR-199a-5p is actively involved in tumor invasion and metastasis as well as in the decline of breast cancer tissues. In this research, overexpression of miR-199a-5p weakened motility and invasion of breast cancer cells MCF-7 and MDA-MB-231. Upregulation of Ets-1 increased breast cancer cell invasion, but the mechanism by which miR-199a-5p modulates activation of Ets-1 in breast cancer was not clarified. We investigated the relationship between miR-199a-5p and Ets-1 on the basis of 158 primary breast cancer case specimens, and the results showed that Ets-1 expression was inversely correlated with endogenous miR-199a-5p. Overexpression of miR-199a-5p reduced the mRNA and protein levels of Ets-1 in MCF-7 and MDA-MB-231 cells, whereas anti-miR-199a-5p elevated Ets-1. siRNA-mediated Ets-1 knockdown phenocopied the inhibition invasion of miR-199a-5p in vitro. Moreover, luciferase reporter assay revealed that miR-199a-5p directly targeted 3'-UTR of Ets-1 mRNA. This research revealed that miR-199a-5p could descend the levels of ß1 integrin by targeting 3'-UTR of Ets-1 to alleviate the invasion of breast cancer via FAK/Src/Akt/mTOR signaling pathway. Our results provide insight into the regulation of ß1 integrin through miR-199a-5p-mediated Ets-1 silence and will help in designing new therapeutic strategies to inhibit signal pathways induced by miR-199a-5p in breast cancer invasion.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Integrina beta1/genética , MicroRNAs/genética , Invasividade Neoplásica/genética , Proteína Proto-Oncogênica c-ets-1/genética , Proteína Proto-Oncogênica c-ets-1/metabolismo , Regiões 3' não Traduzidas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação para Baixo/genética , Feminino , Quinase 1 de Adesão Focal/metabolismo , Humanos , Integrina beta1/biossíntese , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteína Proto-Oncogênica c-ets-1/deficiência , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Transcrição Gênica/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
17.
Int J Oncol ; 49(1): 381-92, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27121503

RESUMO

NF-κB subunits play important roles in carcinogenesis of a variety of human malignancies and response to cancer therapy; however, the contribution of an individual subunit has not been thoroughly defined. Constitutive activation of the canonical NF-κB subunit is a critical event in prostate carcinogenesis. Recent findings point out that RelB, which contributes to the non-canonical NF-κB activity, functions importantly in the prostate cancer progression. Here, we investigated systemically the functional roles of RelB in prostate cancer and examine its significance as a therapeutic target. Targeting RelB using short hairpin RNA approach in androgen-independent DU145 prostate cancer cells interfered with various biological behaviors of cells. We observed that RelB knockdown inhibited prostate cancer cell growth, migration, and invasion, and enhanced proteasome inhibitor sensitivity. The altered expression of anti-apoptotic gene Bcl-2 played critical roles in regulating both spontaneous and radiation-induced apoptosis in the presence of RelB knockdown. For the first time, we showed that RelB knockdown significantly attenuated the migration and invasion of DU145 prostate cancer cells, due to the reduction of integrin ß-1. Collectively, we provided evidence that RelB functioned as an oncogene in prostate cancer. Developing a RelB-targeted therapeutic intervention, is valuable in treating advanced, metastatic prostate cancer.


Assuntos
Integrina beta1/biossíntese , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Fator de Transcrição RelB/genética , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Masculino , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , Inibidores de Proteassoma/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Interferente Pequeno/genética , Tolerância a Radiação/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Fator de Transcrição RelB/biossíntese
18.
Eur Arch Otorhinolaryngol ; 273(4): 845-51, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25864182

RESUMO

E-cadherin, ß-catenin, and ß1 integrin are important cell adhesion molecules to maintain epithelial structure and function. We investigated the expression of these cell adhesion molecules in cholesteatomas to understand the role of cell-cell and cell-extracellular matrix interaction in cholesteatomas. An immunohistochemical investigation was carried out on 35 cholesteatoma tissue samples (14 congenital, 21 acquired cholesteatomas) and 10 normal retroauricular skin (RAS) tissues which are obtained during middle ear surgery. The expression rate was measured to find out differences between retroauricular skin and cholesteatoma, as well as between congenital and acquired cholesteatoma. E-cadherin expression rate was significantly lower in the cholesteatoma (spinous layer 88.7 ± 17.9 %, granular layer 54.6 ± 22.6 %) than in the RAS (100 %, 74.4 ± 7.4 %) and in the acquired (83.3 ± 19.4 %, 48.1 ± 22.9 %) than in the congenital (96.7 ± 12.0 %, 64.4 ± 18.8 %). ß-catenin expression rate was significantly lower in the cholesteatoma (spinous layer 84.1 ± 17.2 %, granular layer 28.7 ± 30.8 %) than in the RAS (100 %, 75.9 ± 6.1 %) and in the acquired (78.1 ± 17.0 %, 17.1 ± 22.3 %) than in the congenital (93.2 ± 13.5 %, 46.1 ± 34.2 %). The expression pattern of ß-catenin is similar to that of E-cadherin. In ß1 integrin, there was no significant difference of the expression rate between RAS and cholesteatoma, as well as between congenital and acquired cholesteatoma. In conclusion, the expression of E-cadherin and ß-catenin is reduced in cholesteatoma, and the reduction is more pronounced in acquired cholesteatoma than in congenital cholesteatoma. Acquired cholesteatomas showed more aggressive characteristics than congenital cholesteatomas in terms of cell-cell adhesion.


Assuntos
Caderinas/genética , Colesteatoma da Orelha Média/genética , Regulação da Expressão Gênica , Integrina beta1/genética , RNA/genética , beta Catenina/genética , Adolescente , Adulto , Idoso , Caderinas/biossíntese , Moléculas de Adesão Celular , Criança , Pré-Escolar , Colesteatoma/congênito , Colesteatoma/genética , Colesteatoma/metabolismo , Colesteatoma/patologia , Colesteatoma da Orelha Média/metabolismo , Colesteatoma da Orelha Média/patologia , Feminino , Humanos , Imuno-Histoquímica , Lactente , Integrina beta1/biossíntese , Masculino , Pessoa de Meia-Idade , Adulto Jovem , beta Catenina/biossíntese
19.
J Biomed Mater Res B Appl Biomater ; 104(6): 1056-63, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26013977

RESUMO

Native cells live in a three-dimensional (3D) extracellular matrix (ECM) capable of regulating cell activities through various physical and chemical factors. Designed topographies have been well proven to trigger significant difference in cell behaviours. However, present topographies are almost all constructed on two-dimensional (2D) substrates like discs and films, which are far from features like 3D and porosity required in application like bone repair. Here we bottom-up assembled poly(lactic-co-glycolic acid)/calcium carbonate (PLGA/CC) microspheres with superficial porous topography intactly into a 3D porous scaffold. Because the scaffold was obtained through a mild technique, the bioactivity of released BMP-2 was well retained. Mouse bone marrow mesenchymal stem cells (mMSCs) were cultured on produced scaffolds having different 3D topographies. It turned out that osteogenic differentiation of mMSCs did respond to the 3D topographies, while proliferation didn't. Gene expression of αv and ß1 integrins revealed that adhesion was supposed to be the underlying mechanism for osteogenic response. The study provides insight into enhancing function of practical scaffolds by elaborate topography design. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 104B: 1056-1063, 2016.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular , Matriz Extracelular/química , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Alicerces Teciduais/química , Animais , Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Integrina alfaV/biossíntese , Integrina beta1/biossíntese , Ácido Láctico/química , Células-Tronco Mesenquimais/citologia , Camundongos , Microesferas , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Porosidade
20.
Carcinogenesis ; 36(9): 1071-83, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26153023

RESUMO

Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin ß1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.


Assuntos
Apoptose/efeitos dos fármacos , Artemisininas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Rabdomiossarcoma Embrionário/tratamento farmacológico , Rabdomiossarcoma Embrionário/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilcisteína/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/fisiologia , Artemisininas/uso terapêutico , Artesunato , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Dano ao DNA/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Integrina beta1/biossíntese , Camundongos , MicroRNAs/biossíntese , Invasividade Neoplásica , Transplante de Neoplasias , Moléculas de Adesão de Célula Nervosa/biossíntese , Fator de Transcrição PAX7/biossíntese , Piridinas/farmacologia , Transplante Heterólogo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA