Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Carbohydr Polym ; 278: 118944, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-34973762

RESUMO

Klebsiella pneumoniae serotype KN2 is a carbapenem-resistant strain and leads to the health care-associated infections, such as bloodstream infections. Its capsular polysaccharide (CPS) was isolated and cleaved by a specific enzyme from a bacteriophage into a hexasaccharide-repeating unit. With GC-MS, NMR, and Mass analyses, the structure of KN2 CPS was determined to be {→3)-ß-D-Glcp-(1→3)-[α-D-GlcpA-(1→4)-ß-D-Glcp-(1→6)]-α-D-Galp-(1→6)-ß-D-Galp-(1→3)-ß-D-Galp-(1→}n. We demonstrated that 1 µg/mL CPS could stimulate J774A.1 murine macrophages to release tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in vitro. Also, we proved that KN2 CPS induced the immune response through Toll-like receptor 4 (TLR4) in the human embryonic kidney (HEK)-293 cells. Strikingly, the hexasaccharide alone shows the same immune response as the CPS, suggesting that the hexasaccharide can shape the adaptive immunity to be a potential vaccine adjuvant. The glucuronic acid (GlcA) on other polysaccharides can affect the immune response, but the GlcA-reduced KN2 CPS and hexasaccharide still maintain their immunomodulatory activities.


Assuntos
Antibacterianos/farmacologia , Carbapenêmicos/farmacologia , Fatores Imunológicos/farmacologia , Klebsiella pneumoniae/efeitos dos fármacos , Polissacarídeos Bacterianos/farmacologia , Receptor 4 Toll-Like/imunologia , Antibacterianos/química , Carbapenêmicos/química , Células HEK293 , Humanos , Fatores Imunológicos/química , Ligantes , Testes de Sensibilidade Microbiana , Polissacarídeos Bacterianos/química
2.
Front Immunol ; 11: 607564, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424855

RESUMO

Conjugated polyenes are a class of widely occurring natural products with various biological functions. We previously identified 4-hydroxy auxarconjugatin B (4-HAB) as anti-inflammatory agent with an IC50 of ~20 µM. In this study, we synthesized a new anti-inflammatory 4-HAB analogue, F240B, which has an IC50 of less than 1 µM. F240B dose-dependently induced autophagy by increasing autophagic flux, LC3 speck formation and acidic vesicular organelle formation. F240B inhibited NACHT, LRR and PYD domain-containing protein 3 (NLRP3) inflammasome activation through autophagy induction. In a mechanistic study, F240B inhibited interleukin (IL)-1ß (IL-1ß) precursor expression, promoted degradation of NLRP3 and IL-1ß, and reduced mitochondrial membrane integrity loss in an autophagy-dependent manner. Additionally, F240B inhibited apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization and speck formation without affecting the interaction between NLRP3 and ASC or NIMA-related kinase 7 (NEK7) and double-stranded RNA-dependent kinase (PKR). Furthermore, F240B exerted in vivo anti-inflammatory activity by reducing the intraperitoneal influx of neutrophils and the levels of IL-1ß, active caspase-1, IL-6 and monocyte chemoattractant protein-1 (MCP-1) in lavage fluids in a mouse model of uric acid crystal-induced peritonitis. In conclusion, F240B attenuated the NLRP3 inflammasome through autophagy induction and can be developed as an anti-inflammatory agent in the future.


Assuntos
Anti-Inflamatórios/farmacologia , Autofagia/efeitos dos fármacos , Inflamassomos/metabolismo , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Peritonite/prevenção & controle , Animais , Anti-Inflamatórios/síntese química , Proteínas Relacionadas à Autofagia/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Peritonite/induzido quimicamente , Peritonite/metabolismo , Peritonite/patologia , Estabilidade Proteica , Células RAW 264.7 , Transdução de Sinais , Células THP-1 , Ácido Úrico
3.
PLoS One ; 9(7): e101679, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25007054

RESUMO

RASSF1C is a major isoform of the RASSF1 gene, and is emerging as an oncogene. This is in contradistinction to the RASSF1A isoform, which is an established tumor suppressor. We have previously shown that RASSF1C promotes lung cancer cell proliferation and have identified RASSF1C target genes with growth promoting functions. Here, we further report that RASSF1C promotes lung cancer cell migration and enhances lung cancer cell tumor sphere formation. We also show that RASSF1C over-expression reduces the inhibitory effects of the anti-cancer agent, betulinic acid (BA), on lung cancer cell proliferation. In previous work, we demonstrated that RASSF1C up-regulates piwil1 gene expression, which is a stem cell self-renewal gene that is over-expressed in several human cancers, including lung cancer. Here, we report on the effects of BA on piwil1 gene expression. Cells treated with BA show decreased piwil1 expression. Also, interaction of IGFBP-5 with RASSF1C appears to prevent RASSF1C from up-regulating PIWIL1 protein levels. These findings suggest that IGFBP-5 may be a negative modulator of RASSF1C/ PIWIL1 growth-promoting activities. In addition, we found that inhibition of the ATM-AMPK pathway up-regulates RASSF1C gene expression.


Assuntos
Proteínas Argonautas/metabolismo , Expressão Gênica , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteínas Supressoras de Tumor/genética , Antineoplásicos/farmacologia , Proteínas Argonautas/genética , Movimento Celular , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Neoplasias Pulmonares , Triterpenos Pentacíclicos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Esferoides Celulares , Triterpenos/farmacologia , Proteínas Supressoras de Tumor/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Ácido Betulínico
4.
Mol Biol Int ; 2013: 145096, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24327924

RESUMO

RASSF1A has been demonstrated to be a tumor suppressor, while RASSF1C is now emerging as a growth promoting protein in breast and lung cancer cells. To further highlight the dual functionality of the RASSF1 gene, we have compared the effects of RASSF1A and RASSF1C on cell proliferation and apoptosis in the presence of TNF- α . Overexpression of RASSF1C in breast and lung cancer cells reduced the effects of TNF- α on cell proliferation, apoptosis, and MST1/2 phosphorylation, while overexpression of RASSF1A had the opposite effect. We also assessed the expression of RASSF1A and RASSF1C in breast and lung tumor and matched normal tissues. We found that RASSF1A mRNA levels are significantly higher than RASSF1C mRNA levels in all normal breast and lung tissues examined. In addition, RASSF1A expression is significantly downregulated in 92% of breast tumors and in 53% of lung tumors. Conversely, RASSF1C was upregulated in 62% of breast tumors and in 47% of lung tumors. Together, these findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor but instead may play a role in stimulating survival in breast and lung cancer cells.

5.
BMC Res Notes ; 5: 239, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22591718

RESUMO

BACKGROUND: RASSF1A and RASSF1C are two major isoforms encoded by the Ras association domain family 1 (RASSF1) gene through alternative promoter selection and mRNA splicing. RASSF1A is a well established tumor suppressor gene. Unlike RASSF1A, RASSF1C appears to have growth promoting actions in lung cancer. In this article, we report on the identification of novel RASSF1C target genes in non small cell lung cancer (NSCLC). METHODS: Over-expression and siRNA techniques were used to alter RASSF1C expression in human lung cancer cells, and Affymetrix-microarray study was conducted using NCI-H1299 cells over-expressing RASSF1C to identify RASSF1C target genes. RESULTS: The microarray study intriguingly shows that RASSF1C modulates the expression of a number of genes that are involved in cancer development, cell growth and proliferation, cell death, and cell cycle. We have validated the expression of some target genes using qRT-PCR. We demonstrate that RASSF1C over-expression increases, and silencing of RASSF1C decreases, the expression of PIWIL1 gene in NSCLC cells using qRT-PCR, immunostaining, and Western blot analysis. We also show that RASSF1C over-expression induces phosphorylation of ERK1/2 in lung cancer cells, and inhibition of the MEK-ERK1/2 pathway suppresses the expression of PIWIL1 gene expression, suggesting that RASSF1C may exert its activities on some target genes such as PIWIL1 through the activation of the MEK-ERK1/2 pathway. Also, PIWIL1 expression is elevated in lung cancer cell lines compared to normal lung epithelial cells. CONCLUSIONS: Taken together, our findings provide significant data to propose a model for investigating the role of RASSF1C/PIWIL1 proteins in initiation and progression of lung cancer.


Assuntos
Proteínas Argonautas/genética , Regulação Neoplásica da Expressão Gênica/genética , Células-Tronco Neoplásicas/metabolismo , Proteínas Supressoras de Tumor/genética , Apoptose/genética , Proteínas Argonautas/metabolismo , Benzamidas/farmacologia , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação/efeitos dos fármacos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor/metabolismo
6.
J Bone Miner Res ; 27(7): 1553-65, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22437732

RESUMO

Claudin 18 (Cldn-18) belongs to a large family of transmembrane proteins that are important components of tight junction strands. Although several claudin members are expressed in bone, the functional role for any claudin member in bone is unknown. Here we demonstrate that disruption of Cldn-18 in mice markedly decreased total body bone mineral density, trabecular bone volume, and cortical thickness in Cldn-18(-/-) mice. Histomorphometric studies revealed that bone resorption parameters were increased significantly in Cldn-18(-/-) mice without changes in bone formation. Serum levels of tartrate-resistant acid phosphatase 5b (TRAP5b) and mRNA expression levels of osteoclast specific markers and signaling molecules were also increased. Loss of Cldn-18 further exacerbated calcium deficiency induced bone loss by influencing bone resorption, thereby resulting in mechanically weaker bone. In vitro studies with bone marrow macrophages revealed Cldn-18 disruption markedly enhanced receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation but not macrophage colony-stimulating factor (MCSF)-induced bone marrow macrophage (BMM) proliferation. Consistent with a direct role for Cldn-18 in regulating osteoclast differentiation, overexpression of wild type but not PDZ binding motif deleted Cldn-18 inhibited RANKL-induced osteoclast differentiation. Furthermore, our findings indicate that Cldn-18 interacts with Zonula occludens 2 (ZO-2) to modulate RANKL signaling in osteoclasts. In conclusion, we demonstrate that Cldn-18 is a novel negative regulator of bone resorption and osteoclast differentiation.


Assuntos
Reabsorção Óssea , Claudinas/biossíntese , Claudinas/fisiologia , Osteoclastos/citologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Proliferação de Células , Feminino , Genótipo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Ligante RANK/metabolismo , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-2
7.
J Gene Med ; 13(10): 511-21, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21898721

RESUMO

BACKGROUND: The present study aimed to develop a rat model of biceps tenodesis and to assess the feasibility of a lentiviral (LV)-based bone morphogenetic protein (BMP) 4 in vivo gene transfer strategy for healing of biceps tenodesis. METHODS: A rat model of biceps tenodesis was developed with an interference-fit open surgical technique. A LV vector expressing a BMP4 gene or ß-galactosidase (ß-gal) control gene was applied to the bone tunnel and the tendon graft before its insertion into the bone tunnel. Osteointegration was assessed by histology and pull-out tensile strength was measured by a biomechanical test suitable for small rat biceps tendon grafts. RESULTS: Neo-chondrogenesis was seen at the tendon-bone interface of LV-BMP4-treated but not control rats. The LV-BMP4-treated rats showed 32% (p < 0.05) more newly-formed trabecular bone at the tendon-bone junction than the LV-ß-gal-treated controls after 3 weeks. However, the sites of neo-chondrogenesis and new bone formation in the LV-BMP4-treated tenodesis were highly spotty. Although the LV-BMP4 strategy did not promote bony integration of the tendon graft, it yielded a 29.5 ± 11.8% (p = 0.066) increase in improvement the pull-out strength of rat biceps tendons compared to the LV-ß-gal treatment after 5 weeks. CONCLUSIONS: Although the LV-BMP4 in vivo gene transfer strategy did not enhance osteointegration of the tendon graft, it yielded a marked improvement in the return of the pull-out strength of the tendon graft. This presumably was largely a result of the bone formation effect of BMP4 that traps or anchors the tendon graft onto the bony tunnel.


Assuntos
Braço/cirurgia , Proteína Morfogenética Óssea 4/genética , Técnicas de Transferência de Genes , Lentivirus/genética , Osseointegração/fisiologia , Tendões/transplante , Tenodese , Animais , Braço/anatomia & histologia , Proteína Morfogenética Óssea 4/metabolismo , Condrogênese/fisiologia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Lentivirus/metabolismo , Masculino , Teste de Materiais , Modelos Animais , Ratos , Ratos Endogâmicos F344 , Procedimentos de Cirurgia Plástica/métodos , Estresse Mecânico , Resistência à Tração
8.
Langmuir ; 27(13): 8424-9, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21630657

RESUMO

We describe a simple and effective method to obtain colloidal surface-functionalized Au nanoparticles. The method is primarily based on irradiation of a gold solution with high-flux X-rays from a synchrotron source in the presence of 11-mercaptoundecanoic acid (MUA). Extensive tests of the products demonstrated high colloidal density as well as excellent stability, shelf life, and biocompatibility. Specific tests with X-ray diffraction, UV-visible spectrometry, visible microscopy, Fourier transform infrared spectroscopy, dark-field visible-light scattering microscopy, and transmission electron microscopy demonstrated that MUA, being an effective surfactant, not only allows tunable size control of the nanoparticles, but also facilitates functionalization. The nanoparticle sizes were 6.45 ± 1.58, 1.83 ± 1.21, 1.52 ± 0.37 and 1.18 ± 0.26 nm with no MUA and with MUA-to-Au ratios of 1:2, 1:1, and 3:1. The MUA additionally enabled functionalization with l-glycine. We thus demonstrated flexibility in controlling the nanoparticle size over a large range with narrow size distribution.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Coloides/química , Coloides/farmacologia , Relação Dose-Resposta a Droga , Ácidos Graxos/química , Ouro/farmacologia , Camundongos , Tamanho da Partícula , Relação Estrutura-Atividade , Compostos de Sulfidrila/química , Propriedades de Superfície
9.
J Gene Med ; 13(2): 77-88, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21322098

RESUMO

BACKGROUND: The present study assesses the effect of the stem cell antigen-1 positive (Sca-1(+) ) cell-based human growth hormone (hGH) ex vivo gene transfer strategy on endosteal bone mass in the mouse. METHODS: Sublethally irradiated recipient mice were transplanted with Sca-1(+) cells transduced with lentiviral vectors expressing hGH or ß-galactosidase control genes. Bone parameters were assessed by micro-computed tomography and histomorphometry. RESULTS: This hGH strategy drastically increased hGH mRNA levels in bone marrow cells and serum insulin-like growth factor-I (IGF-I) (by nearly 50%, p < 0.002) in hGH recipient mice. Femoral trabecular bone volume of the hGH mice was significantly reduced by 35% (p < 0.002). The hGH mice also had decreased trabecular number (by 26%; p < 0.0001), increased trabecular separation (by 38%; p < 0.0002) and reduced trabecular connectivity density (by 64%; p < 0.001), as well as significantly more osteoclasts (2.5-fold; p < 0.05) and greater osteoclastic surface per bone surface (2.6-fold; p < 0.01). CONCLUSIONS: Targeted expression of hGH in cells of marrow cavity through the Sca-1(+) cell-based gene transfer strategy increased circulating IGF-I and decreased endosteal bone mass through an increase in resorption in recipient mice. These results indicate that high local levels of hGH or IGF-I in the bone marrow microenvironment enhanced resorption, which is consistent with previous findings in transgenic mice with targeted bone IGF-I expression showing that high local IGF-I expression increased bone remodeling, favoring a net bone loss. Thus, GH and/or IGF-I would not be an appropriate transgene for use in this Sca-1(+) cell-based gene transfer strategy to promote endosteal bone formation. Published 2011 John Wiley & Sons, Ltd.


Assuntos
Antígenos Ly/metabolismo , Reabsorção Óssea , Técnicas de Transferência de Genes , Hormônio do Crescimento Humano , Proteínas de Membrana/metabolismo , Animais , Antígenos Ly/genética , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/anatomia & histologia , Osso e Ossos/citologia , Osso e Ossos/patologia , Fêmur/anatomia & histologia , Fêmur/citologia , Dosagem de Genes , Regulação da Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células HEK293 , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Radiografia , Transdução Genética , Irradiação Corporal Total
10.
Mol Imaging Biol ; 13(3): 443-451, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20567925

RESUMO

PURPOSE: The purpose of this study was to evaluate the long-term cellular toxicity, labeling efficiency, chondrogenic differentiation capacity, and intracellular distribution following direct superparamagnetic iron oxide (SPIO) nanoparticle labeling of human mesenchymal stem cells (hMSCs) in the absence of transfection agents. PROCEDURES: hMSCs were incubated with a SPIO, Ferucarbotran, at concentrations of 0, 1, 10, and 100 µg Fe/ml for 24 or 72 h. The cell granularity and size change, reactive oxygen species generation, and mitochondria membrane potential were measured by flow cytometry. The differentiation capacity of the cells into chondrocytes was determined by Alcian blue and Safranin-O staining, immunocytochemical analysis, and reverse transcription polymerase chain reaction. RESULTS: The intracellular distribution of the internalized particles was visualized via confocal microscopy. No significant difference was found in the toxicity of labeled cells relative to controls. Successful chondrogenesis of Ferucarbotran-labeled hMSCs was confirmed. The intracellular SPIO nanoparticles were located within the lysosomes. CONCLUSIONS: In conclusion, we have demonstrated the feasibility of direct labeling with Ferucarbotran without impairment of cellular function, toxicity, or inhibition of differentiation capacity. Furthermore, lysosomal metabolism takes place after intracellular uptake of Ferucarbotran.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Dextranos/toxicidade , Espaço Intracelular/metabolismo , Nanopartículas de Magnetita/toxicidade , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Coloração e Rotulagem , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Espaço Intracelular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Microscopia Confocal , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
11.
BMC Cancer ; 10: 562, 2010 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-20955597

RESUMO

BACKGROUND: The Ras association domain family 1 (RASSF1) gene is a Ras effector encoding two major mRNA forms, RASSF1A and RASSF1C, derived by alternative promoter selection and alternative mRNA splicing. RASSF1A is a tumor suppressor gene. However, very little is known about the function of RASSF1C both in normal and transformed cells. METHODS: Gene silencing and over-expression techniques were used to modulate RASSF1C expression in human breast cancer cells. Affymetrix-microarray analysis was performed using T47D cells over-expressing RASSF1C to identify RASSF1C target genes. RT-PCR and western blot techniques were used to validate target gene expression. Cell invasion and apoptosis assays were also performed. RESULTS: In this article, we report the effects of altering RASSF1C expression in human breast cancer cells. We found that silencing RASSF1C mRNA in breast cancer cell lines (MDA-MB231 and T47D) caused a small but significant decrease in cell proliferation. Conversely, inducible over-expression of RASSF1C in breast cancer cells (MDA-MB231 and T47D) resulted in a small increase in cell proliferation. We also report on the identification of novel RASSF1C target genes. RASSF1C down-regulates several pro-apoptotic and tumor suppressor genes and up-regulates several growth promoting genes in breast cancer cells. We further show that down-regulation of caspase 3 via overexpression of RASSF1C reduces breast cancer cells' sensitivity to the apoptosis inducing agent, etoposide. Furthermore, we found that RASSF1C over-expression enhances T47D cell invasion/migration in vitro. CONCLUSION: Together, our findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor, but instead may play a role in stimulating metastasis and survival in breast cancer cells.


Assuntos
Apoptose , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas Supressoras de Tumor/fisiologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Inativação Gênica , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/metabolismo , Resultado do Tratamento , Proteínas Supressoras de Tumor/metabolismo
12.
Nanotechnology ; 21(23): 235103, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20479509

RESUMO

Ultrasmall superparamagnetic iron oxide (USPIO) particles are very useful for cellular magnetic resonance imaging (MRI), which plays a key role in developing successful stem cell therapies. However, their low intracellular labeling efficiency, and biosafety concerns associated with their use, have limited their potential usage. In this study we develop a novel system composed of RBC-derived vesicles (RDVs) for efficient delivery of USPIO particles into human bone marrow mesenchymal stem cells (MSCs) for cellular MRI in vitro and in vivo. RDVs are highly biosafe to their autologous MSCs as manifested by cell viability, differentiation, and gene microarray assays. The data demonstrate the potential of RDVs as intracellular delivery vehicles for biomedical applications.


Assuntos
Compostos Férricos/administração & dosagem , Imageamento por Ressonância Magnética/métodos , Células-Tronco/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Eritrócitos/citologia , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco/metabolismo
13.
Phys Med Biol ; 55(4): 931-45, 2010 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-20090183

RESUMO

Biocompatible Au nanoparticles with surfaces modified by PEG (polyethylene glycol) were developed in view of possible applications for the enhancement of radiotherapy. Such nanoparticles exhibit preferential deposition at tumor sites due to the enhanced permeation and retention (EPR) effect. Here, we systematically studied their effects on EMT-6 and CT26 cell survival rates during irradiation for a dose up to 10 Gy with a commercial biological irradiator (E(average) = 73 keV), a Cu-Kalpha(1) x-ray source (8.048 keV), a monochromatized synchrotron source (6.5 keV), a radio-oncology linear accelerator (6 MeV) and a proton source (3 MeV). The percentage of surviving cells after irradiation was found to decrease by approximately 2-45% in the presence of PEG-Au nanoparticles ([Au] = 400, 500 or 1000 microM). The cell survival rates decreased as a function of the dose for all sources and nanoparticle concentrations. These results could open the way to more effective cancer irradiation therapies by using nanoparticles with optimized surface treatment. Difficulties in applying MTT assays were also brought to light, showing that this approach is not suitable for radiobiology.


Assuntos
Compostos de Ouro , Nanopartículas Metálicas , Polietilenoglicóis , Radioterapia/métodos , Animais , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Coloides/química , Dano ao DNA/efeitos da radiação , Compostos de Ouro/química , Nanopartículas Metálicas/química , Camundongos , Polietilenoglicóis/química , Dosagem Radioterapêutica , Fatores de Tempo
14.
Mol Cell Biol ; 30(3): 711-21, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19995908

RESUMO

Mutations of ephrin B1 in humans result in craniofrontonasal syndrome. Because little is known of the role and mechanism of action of ephrin B1 in bone, we examined the function of osteoblast-produced ephrin B1 in vivo and identified the molecular mechanism by which ephrin B1 reverse signaling regulates bone formation. Targeted deletion of the ephrin B1 gene in type 1alpha2 collagen-producing cells resulted in severe calvarial defects, decreased bone size, bone mineral density, and trabecular bone volume, caused by impairment in osterix expression and osteoblast differentiation. Coimmunoprecipitation of the TAZ complex with TAZ-specific antibody revealed a protein complex containing ephrin B1, PTPN13, NHERF1, and TAZ in bone marrow stromal (BMS) cells. Activation of ephrin B1 reverse signaling with soluble EphB2-Fc led to a time-dependent increase in TAZ dephosphorylation and shuttling from cytoplasm to nucleus. Treatment of BMS cells with exogenous EphB2-Fc resulted in a 4-fold increase in osterix expression as determined by Western blotting. Disruption of TAZ expression using specific lentivirus small hairpin RNA (shRNA) decreased TAZ mRNA by 80% and ephrin B1 reverse signaling-mediated increases in osterix mRNA by 75%. Knockdown of NHERF1 expression reduced basal levels of osterix expression by 90% and abolished ephrin B1-mediated induction of osterix expression. We conclude that locally produced ephrin B1 mediates its effects on osteoblast differentiation by a novel molecular mechanism in which activation of reverse signaling leads to dephosphorylation of TAZ and subsequent release of TAZ from the ephrin B1/NHERF1/TAZ complex to translocate to the nucleus to induce expression of the osterix gene and perhaps other osteoblast differentiation genes. Our findings provide strong evidence that ephrin B1 reverse signaling in osteoblasts is critical for BMS cell differentiation and bone formation.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/genética , Efrina-B1/metabolismo , Osteoblastos/citologia , Osteogênese/genética , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Fatores de Transcrição/metabolismo , Aciltransferases , Animais , Células da Medula Óssea/metabolismo , Efrina-B1/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/metabolismo , Fosfoproteínas/genética , Proteína Tirosina Fosfatase não Receptora Tipo 13/metabolismo , Crânio/anormalidades , Trocadores de Sódio-Hidrogênio/genética , Fator de Transcrição Sp7 , Células Estromais/citologia , Células Estromais/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional
15.
Calcif Tissue Int ; 85(4): 356-67, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19763374

RESUMO

This study evaluated whether the murine leukemia virus (MLV)-based cyclooxygenase-2 (Cox-2) ex vivo gene-transfer strategy promotes healing of calvarial defects and/or synergistically enhances bone morphogenetic protein (BMP) 4-mediated bone regeneration. Gelatin scaffolds impregnated with mouse marrow stromal cells (MSCs) transduced with MLV-expressing BMP4, Cox-2, or a control gene were implanted into mouse calvarial defects. Bone regeneration was assessed by X-ray, dual-energy X-ray absorptiometry, and histology. In vitro, Cox-2 or prostanglandin E(2) enhanced synergistically the osteoblastic differentiation action of BMP4 in mouse MSCs. In vivo, implantation of BMP4-expressing MSCs yielded massive bone regeneration in calvarial defects after 2 weeks, but the Cox-2 strategy surprisingly did not promote bone regeneration even after 4 weeks. Staining for alkaline phosphatase (ALP)-expressing osteoblasts was strong throughout the defect of animals receiving BMP2/4-expressing cells, but defects receiving Cox-2-expressing cells displayed weak ALP staining along the edge of original intact bone, indicating that the Cox-2 strategy lacked bone-regeneration effects. The Cox-2 strategy not only lacked bone-regeneration effects but also suppressed the BMP4-induced bone regeneration. In vitro coculture of Cox-2-expressing MSCs with BMP4-expressing MSCs in gelatin scaffolds reduced BMP4 mRNA transcript levels, suggesting that Cox-2 may promote BMP4 gene silencing in BMP4-expressing cells, which may play a role in the suppressive action of Cox-2 on BMP4-mediated bone formation. In summary, the Cox-2 ex vivo gene-transfer strategy not only lacks bone-regeneration effects but also suppresses the bone-regeneration action of BMP4 in healing of calvarial defects.


Assuntos
Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Regeneração Óssea/fisiologia , Ciclo-Oxigenase 2/metabolismo , Crânio/metabolismo , Células Estromais/metabolismo , Animais , Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/genética , Células Cultivadas , Ciclo-Oxigenase 2/genética , Dinoprostona/metabolismo , Técnicas de Transferência de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , RNA Mensageiro/metabolismo , Crânio/lesões , Células Estromais/citologia
16.
J Gene Med ; 11(10): 877-88, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19629966

RESUMO

BACKGROUND: This study assessed whether a Sca-1+ cell-based ex vivo gene transfer strategy, which has been shown to promote robust endosteal bone formation with a modified fibroblast growth factor-2 (FGF2) gene, can be extended to use with bone morphogenetic protein (BMP)2/4 hybrid gene. METHODS: Sublethally irradiated recipient mice were transplanted with lentiviral (LV)-BMP2/4-transduced Sca-1+ cells. Bone parameters were monitored by pQCT and microCT. Gene expression was assessed by the real-time reverse transcriptase-polymerase chain reaction. RESULTS: Recipient mice of LV-BMP2/4-transduced Sca-1+ cells yielded high engraftment and increased BMP4 mRNA levels in marrow cells; but exhibited only insignificant increases in serum and bone alkaline phosphatase activity compared to control mice. pQCT and microCT analyses of femurs showed that, with the exception of small changes in trabecular bone mineral density and cortical bone mineral content in LV-BMP2/4 mice, there were no differences in measured bone parameters between mice of the LV-BMP2/4 group and controls. The lack of large endosteal bone formation effects with the BMP4 strategy could not be attributed to ineffective engraftment or expansion of BMP4-expressing Sca-1+ cells, an inability of the transduced cells to secrete active BMP4 proteins, or to use of the LV-based vector. CONCLUSIONS: Sca-1+ cell-based BMP4 ex vivo strategy did not promote robust endosteal bone formation, raising the possibility of intrinsic differences between FGF2- and BMP4-based strategies in their ability to promote endosteal bone formation. It emphasizes the importance of choosing an appropriate bone growth factor gene for delivery by this Sca-1+ cell-based ex vivo systemic gene transfer strategy to promote bone formation.


Assuntos
Antígenos Ly/biossíntese , Proteína Morfogenética Óssea 4/biossíntese , Terapia Genética , Proteínas de Membrana/biossíntese , Osteogênese/genética , Osteoporose/terapia , Animais , Proteína Morfogenética Óssea 2/biossíntese , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 4/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Osteoporose/genética , Osteoporose/metabolismo , Proteínas Proto-Oncogênicas c-kit/deficiência , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transdução Genética
17.
Bone ; 44(5): 795-804, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19442627

RESUMO

There is now increasing evidence which suggests an important role for reactive oxygen species (ROS) in the pathogenesis of osteoporosis. However, little is known on the molecular components of the oxidative stress pathway or their functions in bone. In this study, we evaluated the role and mechanism of action of glutaredoxin (Grx) 5, a protein that is highly expressed in bone. Osteoblasts were transfected with Grx5 siRNA and treated with hydrogen peroxide (H(2)O(2)). Grx5 siRNA treatment increased apoptosis while Grx5 overexpression protected MC3T3-E1 cells against H(2)O(2) induced apoptosis and ROS formation. Grx5 deficiency results in impaired biogenesis of Fe-S cluster in yeast. Accordingly, activity of mitochondrial aconitase, whose activity is dependent on Fe-S cluster, decreased in Grx5 siRNA treated cells. Since reduced formation of Fe-S cluster would lead to increased level of free iron, a competitive inhibitor of manganese superoxide dismutase (MnSOD), we measured MnSOD activity in Grx5 deficient osteoblasts and found MnSOD activity was significantly reduced. The consequence of long term inhibition of Grx5 on osteoblast apoptosis was evaluated using lentiviral shRNA technology. Grx5 shRNA cells exhibited higher caspase activity and cardiolipin oxidation in the presence of H(2)O(2). MnSOD activity was rescued by the addition of MnCl(2) to Grx5 shRNA osteoblasts in the presence of H(2)O(2). Our findings are consistent with the hypothesis that Grx5 is an important determinant of osteoblast apoptosis and acts via a molecular pathway that involves regulation of ROS production, cardiolipin oxidation, caspase activity, Fe-S cluster formation, and MnSOD activity.


Assuntos
Apoptose/genética , Glutarredoxinas/fisiologia , Osteoblastos/metabolismo , Estresse Oxidativo/genética , Aconitato Hidratase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Células da Medula Óssea/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo , Superóxido Dismutase/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
18.
Calcif Tissue Int ; 83(3): 202-11, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18709396

RESUMO

LIM mineralization protein-1 (LMP-1) is a novel intracellular osteogenic factor associated with bone development that has been implicated in the bone morphogenetic protein (BMP) pathway. This preliminary study evaluated the possibility of LMP-1-based retroviral gene therapy to stimulate osteoblast differentiation in vitro and fracture repair in vivo. A Moloney leukemia virus (MLV)-based retroviral vector to express LMP-1 with a hemagglutinin (HA) tag was developed, and its effects were evaluated on MC3T3-E1 cell differentiation and in the rat femur fracture model. MC3T3-E1 osteoblasts transduced with the MLV-HA-LMP-1 vector demonstrated significantly increased osteoblast marker gene expression (P < 0.05) and mineral deposition compared to control transduced cells. Femoral midshaft fractures were produced in Fischer 344 rats by the three-point bending technique. The MLV-HA-LMP-1 or control vector was applied at the fracture site through percutaneous injections 1 day postfracture. Analysis of fracture healing of 10 MLV-HA-LMP-1-treated and 10 control MLV-beta-galactosidase (beta-gal)-treated animals was completed at 3 weeks by X-ray, peripheral quantitative computed tomography, and histology. MLV-HA-LMP-1-treated animals had 63% more bone mineral content at the fracture site (P < 0.01), 34% greater total hard callus area (P < 0.05), and 45% less cartilage in the fracture callus (P < 0.05) compared to MLV-beta-gal-treated animals. There was no effect of LMP-1 treatment on the density of the hard callus. Immunohistochemistry revealed expression of the LMP-1 transgene in the fracture callus at 21 days postfracture. Immunohistochemistry also revealed that LMP-1 transgene expression did not result in an increase in BMP-4 expression in the fracture callus. Compared to MLV-BMP-4 gene therapy studies, MLV-HA-LMP-1 gene therapy improved bony union of the fracture gap to a greater extent and did not cause heterotopic bone formation. This suggests that LMP-1 may be a better potential candidate for gene therapy for fracture repair than BMP-4. These exciting, albeit preliminary, findings indicate that LMP-1-based gene therapy may potentially be a simple and effective means to enhance fracture repair that warrants further investigation.


Assuntos
Osso e Ossos/metabolismo , Fraturas Ósseas/terapia , Terapia Genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Osteoblastos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Regeneração Óssea/fisiologia , Osso e Ossos/citologia , Diferenciação Celular , Linhagem Celular , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Proteínas com Domínio LIM , Camundongos , Osteoblastos/citologia , Ratos , Retroviridae , Transdução Genética
19.
J Cell Biochem ; 104(5): 1890-905, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18395833

RESUMO

Insulin-like growth factor binding protein (IGFBP)-6 has been reported to inhibit differentiation of myoblasts and osteoblasts. In the current study, we explored the mechanisms underlying IGFBP-6 effects on osteoblast differentiation. During MC3T3-E1 osteoblast differentiation, we found that IGFBP-6 protein was down-regulated. Overexpression of IGFBP-6 in MC3T3-E1 and human bone cells inhibited nodule formation, osteocalcin mRNA expression and ALP activity. Furthermore, accumulation of IGFBP-6 in the culture media was not required for any of these effects suggesting that IGFBP-6 suppressed osteoblast differentiation by an intracellular mechanism. A yeast two-hybrid screen of an osteosarcoma library was conducted to identify intracellular binding partners to account for IGFBP-6 inhibitory effects on osteoblast differentiation. LIM mineralizing protein (LMP-1) was identified as a high affinity IGFBP-6 binding partner. Physical interaction between IGFBP-6 and LMP-1 was confirmed by co-immunoprecipitation. Fluorescent protein fusion constructs for LMP-1 and IGFBP-6 were transiently transfected into osteoblasts to provide evidence of subcellular locations for each protein. Coexpression of LMP-1-GFP and IGFBP-6-RFP resulted in overlapping subcellular localization of LMP-1 and IGFBP-6. To determine if there was a functional association of IGFBP-6 and LMP-1 as well as a physical association, we studied the effect of IGFBP-6, LMP-1 and their combination on type I procollagen promoter activity. LMP-1 increased promoter activity while IGFBP-6 reduced promoter activity, and coexpression of LMP-1 with IGFBP-6 abrogated IGFBP-6 suppression. These studies provide evidence that overexpression of IGFBP-6 suppresses human and murine osteoblast differentiation, that IGFBP-6 and LMP-1 physically interact, and supports the conclusion that this interaction may be functionally relevant.


Assuntos
Diferenciação Celular , Proteína 6 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Fosfatase Alcalina/metabolismo , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Calcificação Fisiológica , Linhagem Celular , Colágeno Tipo I/genética , Proteínas do Citoesqueleto , Regulação da Expressão Gênica , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/química , Espaço Intracelular/metabolismo , Proteínas com Domínio LIM , Camundongos , Dados de Sequência Molecular , Osteoblastos/enzimologia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Transporte Proteico , Alinhamento de Sequência , Frações Subcelulares/metabolismo
20.
J Gene Med ; 10(3): 229-41, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18088065

RESUMO

BACKGROUND: An in vivo gene therapy strategy was developed to accelerate bone fracture repair. METHODS: Direct injection of a murine leukemia virus-based vector targeted transgene expression to the proliferating periosteal cells arising shortly after fracture. Cyclooxygenase-2 (Cox-2) was selected because the transgene for its prostaglandin products that promote angiogenesis, bone formation and bone resorption, are all required for fracture healing. The human (h) Cox-2 transgene was modified to remove AU-rich elements in the 3'-untranslated region and to improve protein translation. RESULTS: In vitro studies revealed robust and sustained Cox-2 protein expression, prostaglandin E(2) and alkaline phosphatase production in rat bone marrow stromal cells and osteoblasts transgenic for the hCox-2 gene. In vivo studies in the rat femur fracture revealed that Cox-2 transgene expression produced bony union of the fracture by 21 days post-fracture, a time when cartilage persisted within the fracture tissues of control animals and approximately 1 week earlier than the healing normally observed in this model. None of the ectopic bone formation associated with bone morphogenetic protein gene therapy was observed. CONCLUSIONS: This study represents the first demonstration that a single local application of a retroviral vector expressing a single osteoinductive transgene consistently accelerated fracture repair.


Assuntos
Calo Ósseo/metabolismo , Ciclo-Oxigenase 2/genética , Consolidação da Fratura/genética , Terapia Genética/métodos , Retroviridae/genética , Animais , Sequência de Bases , Dinoprostona/biossíntese , Fraturas do Fêmur/genética , Fraturas do Fêmur/terapia , Humanos , Dados de Sequência Molecular , Ratos , Ratos Endogâmicos F344 , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA