Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 112(4): 1161-6, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25587131

RESUMO

T cells are key players in inflammation, autoimmune diseases, and immunotherapy. Thus, holistic and noninvasive in vivo characterizations of the temporal distribution and homing dynamics of lymphocytes in mammals are of special interest. Herein, we show that PET-based T-cell labeling facilitates quantitative, highly sensitive, and holistic monitoring of T-cell homing patterns in vivo. We developed a new T-cell receptor (TCR)-specific labeling approach for the intracellular labeling of mouse T cells. We found that continuous TCR plasma membrane turnover and the endocytosis of the specific (64)Cu-monoclonal antibody (mAb)-TCR complex enables a stable labeling of T cells. The TCR-mAb complex was internalized within 24 h, whereas antigen recognition was not impaired. Harmful effects of the label on the viability, DNA-damage and apoptosis-necrosis induction, could be minimized while yielding a high contrast in in vivo PET images. We were able to follow and quantify the specific homing of systemically applied (64)Cu-labeled chicken ovalbumin (cOVA)-TCR transgenic T cells into the pulmonary and perithymic lymph nodes (LNs) of mice with cOVA-induced airway delayed-type hypersensitivity reaction (DTHR) but not into pulmonary and perithymic LNs of naïve control mice or mice diseased from turkey or pheasant OVA-induced DTHR. Our protocol provides consequent advancements in the detection of small accumulations of immune cells in single LNs and specific homing to the sites of inflammation by PET using the internalization of TCR-specific mAbs as a specific label of T cells. Thus, our labeling approach is applicable to other cells with constant membrane receptor turnover.


Assuntos
Anticorpos Monoclonais/farmacologia , Tomografia por Emissão de Pósitrons/métodos , Receptores de Antígenos/antagonistas & inibidores , Linfócitos T/diagnóstico por imagem , Animais , Apoptose/imunologia , Radioisótopos de Cobre , Dano ao DNA/imunologia , Camundongos , Traçadores Radioativos , Radiografia , Receptores de Antígenos/imunologia , Linfócitos T/imunologia
2.
Magn Reson Med ; 68(5): 1399-409, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22183853

RESUMO

A technique for generation of positive contrast near susceptibility alterations utilizing echo-shifts in k-space is introduced, based on altered Larmor-frequencies and resulting phase-shifts accumulating during the echo-time at the site of local magnetic field gradients. 3D gradient-echo raw-data is acquired and weighted with an inverse Hanning filter. The filter partly suppresses central raw-data points, while maintaining outer areas. Reconstruction of the filtered raw-data results in images where pixels with apparent magnetic field gradients are highlighted against homogeneous pixels. Further processing steps are introduced to remove remaining intensities in the homogeneous parts of the filtered image. Feasibility is shown by an agar phantom containing magnetically labeled cells, with concentrations of 25, 50, 100, and 250 cells/µL, and by images of the human head. The technique allows detection of echo-shifted pixels with automatic suppression of magnetically homogeneous parts while keeping post-processing time short. Fewer than four labeled cells per pixel were clearly displayed with positive contrast. Application to the human head shows bright veins and complete suppression of homogeneous regions. The presented technique has high potential for specific detection of low concentrations of labeled cells or susceptibility altered regions in vivo with positive contrast, whereas areas with low spin density are not highlighted.


Assuntos
Algoritmos , Rastreamento de Células/métodos , Dextranos , Aumento da Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita , Melanoma/patologia , Linhagem Celular Tumoral , Meios de Contraste , Humanos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
3.
Breast Cancer Res ; 13(2): R28, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21392397

RESUMO

INTRODUCTION: Expression of Y-box binding protein-1 (YB-1) is associated with tumor progression and drug resistance. Phosphorylation of YB-1 at serine residue 102 (S102) in response to growth factors is required for its transcriptional activity and is thought to be regulated by cytoplasmic signaling phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways. These pathways can be activated by growth factors and by exposure to ionizing radiation (IR). So far, however, no studies have been conducted on IR-induced YB-1 phosphorylation. METHODS: IR-induced YB-1 phosphorylation in K-RAS wild-type (K-RASwt) and K-RAS-mutated (K-RASmt) breast cancer cell lines was investigated. Using pharmacological inhibitors, small interfering RNA (siRNA) and plasmid-based overexpression approaches, we analyzed pathways involved in YB-1 phosphorylation by IR. Using γ-H2AX foci and standard colony formation assays, we investigated the function of YB-1 in repair of IR-induced DNA double-stranded breaks (DNA-DSB) and postirradiation survival was investigated. RESULTS: The average level of phosphorylation of YB-1 in the breast cancer cell lines SKBr3, MCF-7, HBL100 and MDA-MB-231 was significantly higher than that in normal cells. Exposure to IR and stimulation with erbB1 ligands resulted in phosphorylation of YB-1 in K-RASwt SKBr3, MCF-7 and HBL100 cells, which was shown to be K-Ras-independent. In contrast, lack of YB-1 phosphorylation after stimulation with either IR or erbB1 ligands was observed in K-RASmt MDA-MB-231 cells. Similarly to MDA-MB-231 cells, YB-1 became constitutively phosphorylated in K-RASwt cells following the overexpression of mutated K-RAS, and its phosphorylation was not further enhanced by IR. Phosphorylation of YB-1 as a result of irradiation or K-RAS mutation was dependent on erbB1 and its downstream pathways, PI3K and MAPK/ERK. In K-RASmt cells K-RAS siRNA as well as YB-1 siRNA blocked repair of DNA-DSB. Likewise, YB-1 siRNA increased radiation sensitivity. CONCLUSIONS: IR induces YB-1 phosphorylation. YB-1 phosphorylation induced by oncogenic K-Ras or IR enhances repair of DNA-DSB and postirradiation survival via erbB1 downstream PI3K/Akt and MAPK/ERK signaling pathways.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Genes ras , Proteína 1 de Ligação a Y-Box/metabolismo , Proteína 1 de Ligação a Y-Box/efeitos da radiação , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA , Feminino , Histonas/análise , Humanos , Ligantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Oncogênicas v-erbB/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Tolerância a Radiação/efeitos dos fármacos , Radiação Ionizante , Radiossensibilizantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Proteína 1 de Ligação a Y-Box/genética
4.
J Vasc Interv Radiol ; 22(5): 623-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21414804

RESUMO

PURPOSE: Restenosis is still one of the major limitations after angioplasty. A therapeutic treatment combining ß-irradiation and pharmacologic cyclooxygenase-2 inhibition was employed to study the impact on vascular smooth muscle cells (SMCs). MATERIALS AND METHODS: The effects of meclofenamic acid in combination with yttrium-90 ((90)Y) on cell growth, clonogenic activity, cell migration, and cell cycle distribution of human aortic SMCs were investigated. Treatment was sustained over a period of 4 days and recovery of cells was determined until day 20 after initiation. The hypothesis was that there is no difference between control and treated groups. RESULTS: A dose-dependent growth inhibition was observed in single and combined treatment groups for meclofenamic acid and ß-irradiation. Cumulative radiation dosage of 8 Gy completely inhibited colony formation. This was also observed for 200 µM meclofenamic acid alone or in combination with minor ß-irradiation dosages. Results of the migration tests showed also a dose dependency with additive effects of combined therapy. Meclofenamic acid 200 µM alone and with cumulative ß-irradiation dosages resulted in an increased G2/M-phase share. CONCLUSIONS: Incubating human SMCs with meclofenamic acid and (90)Y for a period of 4 d (ie, 1.5 half-life times) resulted in an effective inhibition of smooth muscle cell proliferation, colony formation, and migration.


Assuntos
Arteriopatias Oclusivas/prevenção & controle , Braquiterapia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ácido Meclofenâmico/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/efeitos da radiação , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos da radiação , Radioisótopos de Ítrio , Angioplastia com Balão/efeitos adversos , Arteriopatias Oclusivas/etiologia , Arteriopatias Oclusivas/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Movimento Celular/efeitos dos fármacos , Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Terapia Combinada , Constrição Patológica , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Humanos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Prevenção Secundária , Fatores de Tempo
5.
BMC Cell Biol ; 11: 22, 2010 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-20370915

RESUMO

BACKGROUND: For clinical applications of mesenchymal stem cells (MSCs), labeling and tracking is crucial to evaluate cell distribution and homing. Magnetic resonance imaging (MRI) has been successfully established detecting MSCs labeled with superparamagnetic particles of iron oxide (SPIO). Despite initial reports that labeling of MSCs with SPIO is safe without affecting the MSC's biology, recent studies report on influences of SPIO-labeling on metabolism and function of MSCs. Exposition of cells and tissues to high magnetic fields is the functional principle of MRI. In this study we established innovative labeling protocols for human MSCs using clinically established SPIO in combination with magnetic fields and investigated on functional effects (migration assays, quantification of colony forming units, analyses of gene and protein expression and analyses on the proliferation capacity, the viability and the differentiation potential) of magnetic fields on unlabeled and labeled human MSCs. To evaluate the imaging properties, quantification of the total iron load per cell (TIL), electron microscopy, and MRI at 3.0 T were performed. RESULTS: Human MSCs labeled with SPIO permanently exposed to magnetic fields arranged and grew according to the magnetic flux lines. Exposure of MSCs to magnetic fields after labeling with SPIO significantly enhanced the TIL compared to SPIO labeled MSCs without exposure to magnetic fields resulting in optimized imaging properties (detection limit: 1,000 MSCs). Concerning the TIL and the imaging properties, immediate exposition to magnetic fields after labeling was superior to exposition after 24 h. On functional level, exposition to magnetic fields inhibited the ability of colony formation of labeled MSCs and led to an enhanced expression of lipoprotein lipase and peroxisome proliferator-activated receptor-gamma in labeled MSCs under adipogenic differentiation, and to a reduced expression of alkaline phosphatase in unlabeled MSCs under osteogenic differentiation as detected by qRT-PCR. Moreover, microarray analyses revealed that exposition of labeled MSCs to magnetic fields led to an up regulation of CD93 mRNA and cadherin 7 mRNA and to a down regulation of Zinc finger FYVE domain mRNA. Exposition of unlabeled MSCs to magnetic fields led to an up regulation of CD93 mRNA, lipocalin 6 mRNA, sialic acid acetylesterase mRNA, and olfactory receptor mRNA and to a down regulation of ubiquilin 1 mRNA. No influence of the exposition to magnetic fields could be observed on the migration capacity, the viability, the proliferation rate and the chondrogenic differentiation capacity of labeled or unlabeled MSCs. CONCLUSIONS: In our study an innovative labeling protocol for tracking MSCs by MRI using SPIO in combination with magnetic fields was established. Both, SPIO and the static magnetic field were identified as independent factors which affect the functional biology of human MSCs. Further in vivo investigations are needed to elucidate the molecular mechanisms of the interaction of magnetic fields with stem cell biology.


Assuntos
Óxido Ferroso-Férrico , Magnetismo , Células-Tronco Mesenquimais/citologia , Nanopartículas , Diferenciação Celular , Proliferação de Células , Humanos , Imageamento por Ressonância Magnética
6.
Magn Reson Med ; 64(4): 1027-38, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20564596

RESUMO

Superparamagnetic iron oxide particles can be utilized to label cells for immune cell and stem cell therapy. The labeled cells cause significant field distortions induced in their vicinity, which can be detected with magnetic resonance imaging (MRI). In conventional imaging, the signal voids arising from the field distortions lead to negative contrast, which is not desirable, as detection of the cells can be masked by native low signal tissue. In this work, a new method for visualizing magnetically labeled cells with positive contrast is proposed and described. The technique presented is based on the susceptibility-weighted imaging (SWI) post-processing algorithm. Phase images from gradient-echo sequences are evaluated pixel by pixel, and a mask is created with values ranging from 0 to 1, depending on the phase value of the pixel. The magnitude image is then multiplied by the mask. With an appropriate mask function, positive contrast in the vicinity of the labeled cells is created. The feasibility of this technique is proved using an agar phantom containing superparamagnetic iron oxide particles-labeled cells and an ex vivo bovine liver. The results show high potential for detecting even small labeled cell concentrations in structurally inhomogeneous tissue types.


Assuntos
Rastreamento de Células/métodos , Hepatócitos/citologia , Fígado/citologia , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita , Animais , Bovinos , Meios de Contraste , Estudos de Viabilidade , Imageamento por Ressonância Magnética/instrumentação , Imagens de Fantasmas , Coloração e Rotulagem/métodos
7.
Cytotherapy ; 11(1): 68-78, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19191056

RESUMO

BACKGROUND AIMS: Labeling of stem cells is crucial to allow tracking of stem cell homing and engraftment after transplantation. In this study we evaluated the influence of cell labeling procedures using clinically approved small particles of iron oxide (SPIO) with or without transfection reagents (TA) on functional parameters of human mesenchymal stem cells (MSC). METHODS: The study was approved by the institutional review board of the University of Tubingen, Germany. Seven populations of bone marrow (BM)-derived human mesenchymal stem cells (MSC) were labeled with SPIO alone or in combination with various TA. Directly after labeling and two passages after labeling migration assays, quantification of colony-forming units and quantitative evaluation of the differentiation potential were performed. Quantification of the cellular total iron load (TIL), determination of the cellular viability and electron microscopy were also performed. RESULTS: Labeling of mesenchymal stem cells with SPIO with or without TA did not affect cell viability and differentiation potential significantly. SPIO in combination with TA coated the cellular surface directly after labeling but was incorporated into the cells after two passages. Labeling of mesenchymal stem cells with TA led to a significant decrease of migration capacity. This effect was abolished after two passages. Labeling with and without TA led to a significant decrease in colony formation ability. This effect could also be observed after two passages. CONCLUSIONS: The observed decrease of migration capacity and colony-formation ability was not associated with either TIL or localization of particles of iron oxide. SPIO labeling with and without TA had functional effects on human mesenchymal stem cells by decreasing the migration capacity and colony-formation ability of the stem cells.


Assuntos
Movimento Celular/efeitos dos fármacos , Meios de Contraste/farmacologia , Ferro/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Óxidos/farmacologia , Coloração e Rotulagem/métodos , Adolescente , Adulto , Idoso , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Dextranos , Feminino , Óxido Ferroso-Férrico , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Nanopartículas de Magnetita , Masculino , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Adulto Jovem
8.
Nucl Med Biol ; 36(2): 163-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19217528

RESUMO

INTRODUCTION: The nucleoside analogue [(18)F]fluorothymidine (FLT) has been designed as a marker of cell proliferation that can be imaged in vivo by positron emission tomography. Clinical pilot studies have demonstrated decreasing FLT uptake following antiproliferative chemotherapy of breast cancer. However, the significance of posttreatment FLT uptake has not been evaluated at the cell level. The aim of this study was to investigate whether FLT uptake detects proliferation inhibition induced by docetaxel or doxorubicin treatment in an in vitro breast cancer model. METHODS: Breast cancer cells (MCF-7) were treated with docetaxel or doxorubicin for 24 h at drug doses inducing 25-99% inhibition of clonogenic survival (IC(25) to IC(99)). Cellular FLT uptake was estimated at 4 h and at 1, 3 and 5 days interval from chemotherapy. [(3)H]Thymidine incorporation and S-phase fraction were measured for comparison. Analysis of variance and the Bland-Altman difference plot were employed for statistical analysis. RESULTS: After treatment, FLT uptake was declined in dependence of the proliferation inhibition mediated by both chemotherapeutic agents (all P<.0001). The decrease of FLT was greater after doxorubicin treatment than after the corresponding docetaxel dose. With doxorubicin (IC(99)), FLT accumulation was reduced by 70% as early as 4 h after treatment. FLT uptake was closely correlated to [(3)H]thymidine incorporation and S-phase fraction (r=.84 to .93). CONCLUSIONS: Right after docetaxel or doxorubicin treatment, FLT uptake corresponds to the reduction of tumor cell proliferation induced. [(18)F]FLT appears promising for monitoring chemosensitivity in breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/farmacologia , Radioisótopos de Flúor , Compostos Radiofarmacêuticos , Taxoides/farmacologia , Timidina/farmacocinética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Docetaxel , Feminino , Humanos , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Fase S
9.
Mol Cancer Ther ; 7(7): 1772-81, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18644989

RESUMO

We have already reported that epidermal growth factor receptor/phosphatidylinositol 3-kinase/AKT signaling is an important pathway in regulating radiation sensitivity and DNA double-strand break (DNA-dsb) repair of human tumor cells. In the present study, we investigated the effect of AKT1 on DNA-dependent protein kinase catalytic subunit (DNA-PKcs) activity and DNA-dsb repair in irradiated non-small cell lung cancer cell lines A549 and H460. Treatment of cells with the specific AKT pathway inhibitor API-59 CJ-OH (API; 1-5 micromol/L) reduced clonogenic survival between 40% and 85% and enhanced radiation sensitivity of both cell lines significantly. As indicated by fluorescence-activated cell sorting analysis (sub-G(1) cells) and poly(ADP-ribose) polymerase cleavage, API treatment or transfection with AKT1-small interfering RNA (siRNA) induced apoptosis of H460 but not of A549 cells. However, in either apoptosis-resistant A549 or apoptosis-sensitive H460 cells, API and/or AKT1-siRNA did not enhance poly(ADP-ribose) polymerase cleavage and apoptosis following irradiation. Pretreatment of cells with API or transfection with AKT1-siRNA strongly inhibited radiation-induced phosphorylation of DNA-PKcs at T2609 and S2056 as well as repair of DNA-dsb as measured by the gamma-H2AX foci assay. Coimmunoprecipitation experiments showed a complex formation of activated AKT and DNA-PKcs, supporting the assumption that AKT plays an important regulatory role in the activation of DNA-PKcs in irradiated cells. Thus, targeting of AKT enhances radiation sensitivity of lung cancer cell lines A549 and H460 most likely through specific inhibition of DNA-PKcs-dependent DNA-dsb repair but not through enhancement of radiation-induced apoptosis.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteína Quinase Ativada por DNA/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Tolerância a Radiação , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Mutação/genética , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Poli(ADP-Ribose) Polimerases/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/efeitos da radiação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/efeitos da radiação , Radiação Ionizante , Proteínas ras/genética
10.
Mol Cancer ; 7: 69, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18789131

RESUMO

BACKGROUND: To elucidate the role of src kinase in caveolin-1 driven internalization and nuclear transport of EGFR linked to regulation of DNA-repair in irradiated cells. RESULTS: Ionizing radiation resulted in src kinase stabilization, activation and subsequent src mediated caveolin-1 Y14- and EGFR Y845-phosphorylations. Both phosphorylations were radiation specific and could not be observed after treatment with EGF. Inhibition of EGFR by the antibody Erbitux resulted in a strong accumulation of caveolin/EGFR complexes within the cytoplasm, which could not be further increased by irradiation. Radiation-induced caveolin-1- and EGFR-phosphorylations were associated with nuclear EGFR transport and activation of DNA-PK, as detected by phosphorylation at T2609. Blockage of src activity by the specific inhibitor PP2, decreased nuclear transport of EGFR and inhibited caveolin-1- and DNA-PK-phosphorylation. Knockdown of src by specific siRNA blocked EGFR phosphorylation at Y845, phosphorylation of caveolin-1 at Y14 and abolished EGFR transport into the nucleus and phosphorylation of DNA-PK. Consequently, both knockdown of src by specific siRNA and also inhibition of src activity by PP2 resulted in an enhanced residual DNA-damage as quantified 24 h after irradiation and increased radiosensitivity. CONCLUSION: Src kinase activation following irradiation triggered caveolin-1 dependent EGFR internalization into caveolae. Subsequently EGFR shuttled into the nucleus. As a consequence, inhibition of internalization and nuclear transport of EGFR blocked radiation-induced phosphorylation of DNA-PK and hampered repair of radiation-induced double strand breaks.


Assuntos
Caveolina 1/metabolismo , Núcleo Celular/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , Receptores ErbB/metabolismo , Transporte Ativo do Núcleo Celular/efeitos da radiação , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Cetuximab , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Humanos , Fosforilação/efeitos da radiação , RNA Interferente Pequeno/metabolismo , Transfecção , Quinases da Família src/metabolismo
11.
Radiother Oncol ; 86(3): 375-82, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18237807

RESUMO

BACKGROUND AND PURPOSE: The purpose of the study was to elucidate the underlying molecular mechanism of the radioprotector, Bowman-Birk proteinase inhibitor (BBI), and its interaction with EGFR nuclear transport. MATERIALS AND METHODS: Molecular effects of BBI at the level of EGFR responses were investigated in vitro with wt. TP53 bronchial carcinoma cell line A549 and the transformed fibroblast cell line HH4dd characterized by a mt. TP53. EGFR and associated protein expression were quantified by Western blotting and confocal microscopy in the cytoplasmic and nuclear cell fraction. Residual DNA double strand breaks were quantified by means of a gammaH(2)AX focus assay. RESULTS: Both irradiation and BBI-treatment stimulated EGFR internalization into the cytoplasm. This process involved src kinase activation, EGFR phosphorylation at Y845, and caveolin 1 phosphorylation at Y14. EGFR internalization correlated with nuclear EGFR transport and was associated with phosphorylation of EGFR at T654. Nuclear EGFR was linked with DNA-PK complex formation and activation. Furthermore, nuclear EGFR was found in complex with TP53, phosphorylated at S15, and with MDC1, following irradiation and BBI treatment. It is noteworthy that MDC1 was strongly decreased in the nuclear EGFR complex in cells with mt. TP53 and failed to be increased by either BBI treatment or irradiation. Interestingly, in cells with mt. TP53 the BBI mediated stimulation of double strand break repair was hampered significantly. CONCLUSION: These data indicate that BBI stimulates complex formation between EGFR, TP53 and MDC1 protein in wt. TP53 cells only. Since MDC1 is essential for recruitment of DNA repair foci, this observation may explain how BBI selectively stimulated repair of DNA double strand breaks in wt. TP53 cells.


Assuntos
Reparo do DNA/efeitos dos fármacos , Receptores ErbB/metabolismo , Inibidores de Serina Proteinase/farmacologia , Inibidor da Tripsina de Soja de Bowman-Birk/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Western Blotting , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Expressão Gênica , Humanos , Microscopia Confocal , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Transativadores/metabolismo , Proteína Supressora de Tumor p53/metabolismo
12.
Radiother Oncol ; 86(3): 383-90, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18037521

RESUMO

PURPOSE: To elucidate the interaction between radioprotector O-phospho-l-tyrosine (P-Tyr) with epidermal growth factor receptor (EGFR). METHODS: Molecular effects of P-Tyr at the level of EGFR responses were investigated in vitro with TP53-wildtype bronchial carcinoma cell line A549, which is radio-protected by P-Tyr treatment. Nuclear EGFR accumulation was followed by confocal microscopy and Western blotting. PKCepsilon protein expression was impaired by specific siRNA. Residual DNA-damage was quantified with gammaH(2)AX foci analysis. RESULTS: P-Tyr mediated radio-protection was associated with nuclear EGFR accumulation. Radiation-induced nuclear EGFR presented increased phosphorylation at residue No. T654. We identified PKCepsilon as responsible for T654-phosphorylation. Knockdown of PKCepsilon by siRNA blocked both radiation- and P-Tyr-triggered nuclear EGFR accumulation. Furthermore, nuclear accumulation of EGFR was associated with increased phosphorylation of DNA-dependent protein kinase (DNA-PK) at residue No. T2609, essential for DNA-repair. Consequently P-Tyr mediated effects upon DNA-PK resulted in a significant reduction of radiation-induced residual gammaH(2)AX-foci. Knockdown of PKCepsilon increased radiation-induced residual damage and abolished the P-Tyr associated radioprotection. In addition, P-Tyr mediated radioprotection was completely absent in colony formation assay. CONCLUSION: The data presented herein suggest that P-Tyr-treatment mediates activation of PKCepsilon, which triggers nuclear EGFR accumulation. Nuclear EGFR is involved in phosphorylation of DNA-PK at Thr2609, which has a significant impact upon DNA-DSB repair.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA/fisiologia , Receptores ErbB/metabolismo , Proteína Quinase C-épsilon/metabolismo , Protetores contra Radiação , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Microscopia Confocal , Fosfotirosina/farmacologia
13.
Cell Transplant ; 17(8): 923-41, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19069635

RESUMO

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS). Mesenchymal stem cells (MSC) have been shown to ameliorate symptoms in experimental autoimmune encephalomyelitis (EAE), a model of MS. Using cloned MSC labeled with clinically approved small particles of iron oxide (SPIO) for treatment of EAE we analyzed the tissue localization of transferred cells. Treatment with unlabeled MSC led to disease amelioration compared to controls. In contrast, treatment with SPIO-labeled MSC lead to increase in disease severity. Treatment with SPIO alone did not alter disease course. After transplantation labeled and nonlabeled MSC were detected in the CNS and the liver with significantly more SPIO-labeled cells present in the CNS. Iron deposition was present in the group treated with SPIO-labeled MSC, indicating that in vivo the initially cell surface-bound iron detached from the MSC. These results could be of great importance for imaging of patients in the clinical setting, indicating that in vivo application of SPIO-labeled MSC needs to be performed with caution because the cell-derived exposure of iron can lead to disease aggravation.


Assuntos
Corantes/efeitos adversos , Encefalomielite Autoimune Experimental/cirurgia , Compostos Férricos/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Coloração e Rotulagem/métodos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/citologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiopatologia , Corantes/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Compostos Férricos/metabolismo , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/fisiologia , Fígado/citologia , Fígado/patologia , Fígado/fisiopatologia , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Microscopia Eletrônica de Transmissão , Esclerose Múltipla/cirurgia , Ratos
14.
Radiother Oncol ; 84(3): 328-34, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17714814

RESUMO

BACKGROUND AND PURPOSE: Purpose of the study was to elucidate the underlying molecular mechanism of the radioprotector O-phospho-tyrosine (P-Tyr). METHODS: Molecular effects of P-Tyr at the level of EGFR responses were investigated in vitro with bronchial carcinoma cell line A549. Nuclear EGFR transport and DNA-PK activation were quantified after Western blotting. Residual DNA-damages were quantified by help of gammaH(2)AX focus assay. RESULTS: As determined by dose-response curves, treatment of cells with P-Tyr for 16h before irradiation results in radioprotection. Simultaneous treatment with EGFR blocking antibody Cetuximab abolished P-Tyr associated radioprotection. At the molecular level P-Tyr mediated a general phosphorylation of EGFR and a pronounced phosphorylation of nuclear EGFR at residue Thr No. 654, also observed after treatment with ionizing radiation. This phosphorylation was associated with nuclear EGFR accumulation. Moreover, P-Tyr-triggered EGFR nuclear accumulation was associated with phosphorylation of DNA-PK at Thr 2609. This activated form of DNA-PK was not DNA associated, but after radiation, DNA binding increased, particularly after P-Tyr pre-treatment. These molecular effects of P-Tyr resulted in a reduction of residual DNA-damage after irradiation. CONCLUSIONS: Radioprotection by P-Tyr is mediated through its stimulation of nuclear EGFR transport and concurrent, but DNA-damage independent, activation of DNA-PK. Thus, subsequent irradiation results in increased binding of DNA-PK to DNA, improved DNA-repair and increased cell survival.


Assuntos
Reparo do DNA/fisiologia , Proteína Quinase Ativada por DNA/metabolismo , Receptores ErbB/metabolismo , Fosfotirosina/fisiologia , Protetores contra Radiação , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Western Blotting , Carcinoma Broncogênico/metabolismo , Linhagem Celular Tumoral , Cetuximab , Humanos , Neoplasias Pulmonares/metabolismo , Fosforilação , Fosfotirosina/farmacologia
15.
Radiother Oncol ; 82(2): 229-38, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17207548

RESUMO

PURPOSE: To evaluate the potential radiosensitizing effect of the specific COX-2 inhibitor celecoxib (Celebrex) on prostate carcinoma cells in vitro. MATERIALS AND METHODS: The influence of celecoxib (concentration range 5 to 75 microM) on radiation-induced cellular and clonogenic survival was investigated in prostate carcinoma cell lines PC-3, DU145, LNCaP and normal prostate epithelial cells (PrEC). Western blot analysis and ELISA were used to determine the impact of radiation alone or radiation combined with celecoxib treatment on COX-2 expression and prostaglandin E2 synthesis. To evaluate induction of celecoxib-induced apoptosis cell cycle analysis has been performed. RESULTS: Celecoxib (5, 10 and 25 microM) in combination with single-dose irradiation of 2 Gy induced a significant radiosensitization in normal prostate epithelial cells which could not be observed for any of the prostate carcinoma cell lines investigated. Increased COX-2 protein expression in PC-3 cells was obvious only after IR with 15 Gy, while PGE2 production was elevated following irradiation (2-15 Gy) in a dose-dependent manner. Treatment with celecoxib alone or in combination with IR led to a dose-dependent increase in COX-2 protein expression. Nevertheless pre-treatment with celecoxib caused a marked reduction of radiation-induced enzyme activity as tested at the level of PGE2 production, both in PC-3 and DU145 cells. Following fractionated irradiation with single doses of 2 Gy, elevated COX-2 protein expression as well as enhanced PGE2 production was observed already after the second fraction in PC-3 cells. Pre-treatment with celecoxib reduced the amount of PGE(2) significantly, but not of COX-2 protein. CONCLUSIONS: Our data obtained for the human prostate cancer cell lines do not indicate that a marked inhibition of prostaglandin E2 synthesis by celecoxib leads to enhanced radiosensitization. Thus, in terms of radiosensitization the analysed prostate cancer cells can be classified as non-responders to celecoxib treatment.


Assuntos
Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/radioterapia , Pirazóis/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Sulfonamidas/farmacologia , Apoptose , Celecoxib , Ciclo Celular , Dinoprostona/biossíntese , Relação Dose-Resposta à Radiação , Humanos , Masculino , Neoplasias da Próstata/metabolismo
16.
Invest Radiol ; 42(4): 212-7, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17351426

RESUMO

OBJECTIVES: We investigated the influence of magnetic resonance (MR) at 3.0 T on clonogenic ability, proliferation, and cell cycle in an embryonic human cell line. MATERIALS AND METHODS: Cells (human lung fibroblasts Hel 299) were exposed to the static magnetic field (3.0 T) of a magnetic resonance imager (MRI) and to a turbo spin echo sequence at 3.0 T within clinical limitations (specific absorption rate 0.92 W/kg). A special MR-compatible incubation system was used. A control group (sham-exposed) and a MRI group (exposed) were set up. We investigated 3 biologic endpoints: colony forming, cell cycle, and proliferation ability. The exposure time was 2 hours in each experiment. RESULTS: In the statistical analysis, none of these tests showed relevant differences between the exposed and sham-exposed group. CONCLUSIONS: No influences of the static field alone as well as a turbo spin echo sequence at 3.0 T on clonogenic ability, proliferation, or cell cycle in eugenic human lung fibroblasts were found.


Assuntos
Ciclo Celular/fisiologia , Proliferação de Células , Campos Eletromagnéticos/efeitos adversos , Fibroblastos/fisiologia , Pulmão/fisiologia , Imageamento por Ressonância Magnética/instrumentação , Humanos , Técnicas In Vitro , Imageamento por Ressonância Magnética/efeitos adversos , Células-Tronco
17.
Radiother Oncol ; 75(2): 237-45, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15890420

RESUMO

BACKGROUND AND PURPOSE: To elucidate the role of TP53 on differential effects of topoisomerase I inhibitor topotecan (Hycamtin on radiation sensitivity. MATERIALS AND METHODS: Cell cycle distribution and protein expression of TP53, p21(WAF1/CIP1) and cyclin B was studied in CCD32 lung fibroblasts, glioblastoma cell lines U118 (mutant TP53), and U87 (wildtype TP53) after treatment with topotecan (0.05 and 1 microM) and/or ionizing radiation (2 Gy). RESULTS: Cell cycle effects varied with topotecan concentration, resulting in G1 arrest (1 microM), or S/G2/M arrest (0.05 microM), and was modified differentially in fibroblasts and in glioblastoma cells in combination with irradiation. Phosphorylation of TP53 and expression of p21(WAF1/CIP1) was induced by IR and/or topotecan in CCD32 cells, and in U118 cells after topotecan treatment, accompanied by cyclin B degradation. In U87 cells only 1 microM topotecan generated phosphorylation of TP53 and p21(WAF1/CIP1) expression; 0.05 microM caused stabilization of cyclin B. CONCLUSIONS: The antagonistic effect of combined topotecan/irradiation treatment in fibroblasts was most likely due to an immediate radiation induced G1 arrest, but was not observed in p53 wildtype glioblastoma cells. Thus, the impact of TP53 on the topotecan response remains indistinct, and is obviously influenced by other genomic alterations acquired by tumor cells.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Glioblastoma/genética , Glioblastoma/patologia , Topotecan/farmacologia , Proteína Supressora de Tumor p53/biossíntese , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Ciclina B/biossíntese , Ciclina B/genética , Ciclina B/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Relação Dose-Resposta a Droga , Fibroblastos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Pulmão/citologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia
18.
Curr Opin Investig Drugs ; 3(8): 1196-8, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12211414

RESUMO

Asahi Kasei is developing a recombinant thrombomodulin (ART-123), a human protein with both thrombin inhibiting and protein C stimulating activities, for the potential treatment of thromboembolism and blood clotting disorders, such as disseminated intravascular thromboembolism [169907]. By September 2000, it had entered phase III trials [383525]. A patent, US-05916874, claiming a method for treating liver injury and whose composition comprises a thrombomodulin, was published in 1999 by the company.


Assuntos
Transtornos da Coagulação Sanguínea/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Tromboembolia/tratamento farmacológico , Trombomodulina/uso terapêutico , Animais , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Humanos , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Trombomodulina/metabolismo
19.
Invest Radiol ; 37(5): 248-53, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11979150

RESUMO

RATIONALE AND OBJECTIVES: The aim of the study was to examine the effects of azelastine on proliferation, clonogenic activity, cell-cycle, and migration of human aortic smooth-muscle cells (haSMCs) in vitro. METHODS: HaSMCs were treated for 4 days with azelastine (1 micromol/L, 25 micromol/L, 50 micromol/L). Half of the treated groups were incubated again with azelastine, the other half received azelastine-free medium every 4 days until day 20. The growth kinetics and clonogenic activity were assessed. The cell-cycle distribution was investigated by FACS -- analysis and the migratory ability was evaluated. RESULTS: Azelastine inhibited the proliferation and the clonogenic activity of haSMCs in a dose dependent manner. A G2/M-phase block was induced and the migratory ability was significantly impaired. CONCLUSION: Azelastine has the potential to inhibit the proliferation of haSMCs. If a sufficient dose can be applied either systemically or locally it could be a valuable substance to prevent restenosis.


Assuntos
Antialérgicos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Ftalazinas/farmacologia , Aorta/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Técnicas In Vitro , Fatores de Tempo
20.
Invest Radiol ; 39(9): 565-71, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15308939

RESUMO

PURPOSE: We sought to evaluate the growth-modulating potential of paclitaxel on cultured human arterial smooth muscle cells depending on the administered dose. MATERIAL AND METHODS: For all experiments human arterial smooth muscle cells (SMCs) were used. SMCs were either cultured for 5 days or for 20 days with paclitaxel (doses: 10(-7) M, 10(-8) M, 10(-9) M). For a total period of 20 days, proliferation kinetics of the SMC were analyzed. To assess the clonogenic activity of the SMC colony-forming assays were performed. Drug- and dose-dependent cell cycle changes were analyzed by flow cytometry. The effect on cell migration was examined in a 2-chamber migration system. The effects of paclitaxel on the synthesis of tenascin were examined via immunofluorescence. RESULTS: Depending on the dose administered, paclitaxel proved to inhibit SMC proliferation effectively when administered during the total period of 20 days. When incubated for 5 days with doses of paclitaxel ranging between 10(-8) M and 10(-9) M, SMCs showed clear signs of regeneration. When being incubated with 10(-7) M of paclitaxel, however, SMCs reacted with a reduction in cell proliferation, a reduced clonogenic activity, and a drug-induced G2/M phase block. SMC migration was inhibited effectively as well as extracellular matrix formation. CONCLUSION: Paclitaxel is a potent inhibitor of SMC proliferation, SMC migration, and extracellular matrix formation in vitro, with all three phases of the restenosis process inhibited effectively.


Assuntos
Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Músculo Liso Vascular/citologia , Paclitaxel/farmacologia , Tenascina/biossíntese , Angioplastia com Balão , Artérias/patologia , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Constrição Patológica , Relação Dose-Resposta a Droga , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Recidiva
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA