Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Imaging ; 152016.
Artigo em Inglês | MEDLINE | ID: mdl-27030399

RESUMO

OBJECTIVES: To use the superparamagnetic iron oxide (SPIO) contrast agent Resovist (±transfection agent) to label human melanoma cells and determine its effects on cellular viability, microstructure, iron quantity, and magnetic resonance imaging (MRI) detectability. MATERIALS AND METHODS: Human SK-Mel28 melanoma cells were incubated with Resovist (±liposomal transfection agent DOSPER). The cellular iron content was measured, and labeled cells were examined at 1.5 T and 3.0 T. The intracellular and extracellular distributions of the contrast agent were assessed by light and electron microscopy. RESULTS: The incubation of melanoma cells with SPIO does not interfere with cell viability or proliferation. The iron is located both intracellularly and extracellularly as iron clusters associated with the exterior of the cell membrane. Despite thorough washing, the extracellular SPIO remained associated with the cell membrane. The liposomal transfection agent does not change the maximum achievable cellular iron content but promotes a faster iron uptake. The MRI detectability persists for at least 7 days. CONCLUSION: The transfection agent DOSPER facilitates the efficient labeling of human metastatic melanoma cells with Resovist. Our findings raise the possibility that other Resovist-labeled cells may collect associated extracellular nanoparticles. The SPIO may be available to other iron-handling cells and not completely compartmentalized during the labeling procedure.


Assuntos
Rastreamento de Células/métodos , Meios de Contraste/farmacologia , Dextranos/farmacologia , Imageamento por Ressonância Magnética/métodos , Melanoma/diagnóstico por imagem , Neoplasias Cutâneas/diagnóstico por imagem , Linhagem Celular Tumoral , Membrana Celular/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ácidos Graxos Monoinsaturados/farmacologia , Humanos , Técnicas In Vitro , Ferro/análise , Nanopartículas de Magnetita , Melanoma/química , Neoplasias Cutâneas/química , Coloração e Rotulagem
2.
NMR Biomed ; 28(8): 1049-58, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26147577

RESUMO

Direct stem cell therapies for functionally impaired tissue require a sufficient number of cells in the target region and a method for verifying the fate of the cells in the subsequent time course. In vivo MRI of iron labeled mesenchymal stem cells has been suggested to comply with these requirements. The study was conducted to evaluate proliferation, migration, differentiation and adhesion effects as well as the obtained iron load of an iron labeling strategy for mesenchymal stem cells. After injection into the porcine urethral sphincter, the labeled cells were monitored for up to six months using MRI. Mesenchymal stem cells were labeled with ferucarbotran (60/100/200 µg/mL) and ferumoxide (200 µg/mL) for the analysis of migration and viability. Phantom MR measurements were made to evaluate effects of iron labeling. For short and long term studies, the iron labeled cells were injected into the porcine urethral sphincter and monitored by MRI. High resolution anatomical images of the porcine urethral sphincter were applied for detection of the iron particles with a turbo-spin-echo sequence and a gradient-echo sequence with multiple TE values. The MR images were then compared with histological staining. The analysis of cell function after iron labeling showed no effects on proliferation or differentiation of the cells. Although the adherence increases with higher iron dose, the ability to migrate decreases as a presumed effect of iron labeling. The iron labeled mesenchymal stem cells were detectable in vivo in MRI and histological staining even six months after injection. Labeling with iron particles and subsequent evaluation with highly resolved three dimensional data acquisition allows sensitive tracking of cells injected into the porcine urethral sphincter for several months without substantial biological effects on mesenchymal stem cells.


Assuntos
Rastreamento de Células/métodos , Ferro , Imageamento por Ressonância Magnética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Uretra/citologia , Animais , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Meios de Contraste , Endoscopia/métodos , Células-Tronco Mesenquimais/fisiologia , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Coloração e Rotulagem/métodos , Suínos , Uretra/cirurgia
3.
J Vasc Interv Radiol ; 26(9): 1388-95, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26169455

RESUMO

PURPOSE: To compare the effects of sirolimus, paclitaxel, and combretastatin A4 (CA4) on regulatory proteins of the cell cycle in proliferating smooth muscle cells (SMCs). MATERIALS AND METHODS: Human aortic SMCs were treated with sirolimus, paclitaxel, and CA4 at 5 × 10(-9) mol/L. After 1 day, half of the cells were harvested (DAY1 group). The treatment medium of the other half was replaced with culture medium on day 4, and those cells were harvested on day 5 (DAY5 group). Cyclins D1, D2, E, and A and cyclin-dependent kinase (CDK) inhibitors p16, p21, and p27 were detected by Western blot technique. Quantification was performed by scanning densitometry of the specific bands. RESULTS: In the DAY1 group, treatment with sirolimus resulted in decreased intracellular levels of cyclins D2 and A (P < .05). Increased D cyclins and reduced levels of cyclins E and A (P < .05) in the DAY5 group indicated a permanent G1/S block by sirolimus. Paclitaxel led to only slight alterations of cyclin and CDK inhibitor expression (P > .05). In the DAY1 group, CA4 decreased intracellular levels of cyclins D2, E, and A (P < .05). Despite recovery effects in the DAY5 group (increase of cyclins D1, D2, and A compared with DAY1 group; P < .05), the upregulation of the CDK inhibitor p21, increased D cyclins, and decreased cyclins E and A (P < .05) are compatible with a G1 arrest. CONCLUSIONS: CA4 is a stronger inhibitor of the SMC cycle than sirolimus or paclitaxel and may represent an alternative for drug-eluting stents in atherosclerotic luminal stenosis. The effect of CA4 on neointima formation should be evaluated further.


Assuntos
Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima/prevenção & controle , Paclitaxel/administração & dosagem , Sirolimo/administração & dosagem , Estilbenos/administração & dosagem , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Neointima/patologia , Resultado do Tratamento , Moduladores de Tubulina/administração & dosagem
4.
J Nucl Med ; 55(2): 301-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24434289

RESUMO

UNLABELLED: Although T cells can be labeled for noninvasive in vivo imaging, little is known about the impact of such labeling on T-cell function, and most imaging methods do not provide holistic information about trafficking kinetics, homing sites, or quantification. METHODS: We developed protocols that minimize the inhibitory effects of (64)Cu-pyruvaldehyde-bis(N4-methylthiosemicarbazone) ((64)Cu-PTSM) labeling on T-cell function and permit the homing patterns of T cells to be followed by PET. Thus, we labeled ovalbumin (OVA) T-cell receptor transgenic interferon (IFN)-γ-producing CD4(+) T (Th1) cells with 0.7-2.2 MBq of (64)Cu-PTSM and analyzed cell viability, IFN-γ production, proliferation, apoptosis, and DNA double-strand breaks and identified intracellular (64)Cu accumulation sites by energy dispersive x-ray analysis. To elucidate the fate of Th1 cell homing by PET, 10(7 64)Cu-OVA-Th1 cells were injected intraperitoneally or intravenously into healthy mice. To test the functional capacities of (64)Cu-OVA-Th1 cells during experimental OVA-induced airway hyperreactivity, we injected 10(7 64)Cu-OVA-Th1 cells intraperitoneally into OVA-immunized or nonimmunized healthy mice, which were challenged with OVA peptide or phosphate-buffered saline or remained untreated. In vivo PET investigations were followed by biodistribution, autoradiography, and fluorescence-activated cell sorting analysis. RESULTS: PET revealed unexpected homing patterns depending on the mode of T-cell administration. Within 20 min after intraperitoneal administration, (64)Cu-OVA-Th1 cells homed to the perithymic lymph nodes (LNs) of naive mice. Interestingly, intravenously administered (64)Cu-OVA-Th1 cells homed predominantly into the lung and spleen but not into the perithymic LNs. The accumulation of (64)Cu-OVA-Th1 cells in the pulmonary LNs (6.8 ± 1.1 percentage injected dose per cubic centimeter [%ID/cm(3)]) 24 h after injection was highest in the OVA-immunized and OVA-challenged OVA airway hyperreactivity-diseased littermates 24 h after intraperitoneal administration and lowest in the untreated littermates (3.7 ± 0.4 %ID/cm(3)). As expected, (64)Cu-OVA-Th1 cells also accumulated significantly in the pulmonary LNs of nonimmunized OVA-challenged animals (6.1 ± 0.5 %ID/cm(3)) when compared with phosphate-buffered saline-challenged animals (4.6 ± 0.5 %ID/cm(3)). CONCLUSION: Our protocol permits the detection of Th1 cells in single LNs and enables temporal in vivo monitoring of T-cell homing over 48 h. This work enables future applications for (64)Cu-PTSM-labeled T cells in clinical trials and novel therapy concepts focusing on T-cell-based immunotherapies of autoimmune diseases or cancer.


Assuntos
Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/diagnóstico por imagem , Compostos Organometálicos , Tomografia por Emissão de Pósitrons/métodos , Células Th1/citologia , Tiossemicarbazonas , Animais , Apoptose , Autoimunidade , Movimento Celular , Proliferação de Células , Separação Celular , Sobrevivência Celular , Radioisótopos de Cobre , Quebras de DNA de Cadeia Dupla , Citometria de Fluxo , Imunoterapia/métodos , Interferon gama/metabolismo , Linfonodos/patologia , Camundongos , Peptídeos/química , Fatores de Tempo , Distribuição Tecidual
5.
Eur Radiol ; 22(9): 1998-2006, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22569995

RESUMO

OBJECTIVES: To investigate the influence of dual antiplatelet therapy vs. aspirin alone on local platelet activation and clinical endpoints in patients with PAD treated with endovascular therapy. METHODS: Patients received either 500 mg aspirin and 300 mg clopidogrel before intervention followed by a daily dose of 100 mg aspirin and 75 mg clopidogrel for 6 months, or the same doses of aspirin plus placebo instead of clopidogrel. Primary endpoints were local concentrations of platelet activation markers ß-thromboglobulin and CD40L, and the rate of patient's resistant to clopidogrel. Secondary endpoints included the clinical development 6 months after the intervention. RESULTS: Eighty patients, 40 in each group, were enrolled. The median peri-interventional concentration of ß-TG was 224.5 vs. 365.5 (P = 0.03) in the clopidogrel and placebo group. The concentration of CD40L was 127 and 206.5 (P = 0.05). Thirty per cent of patients who had received clopidogrel were resistant. Two clopidogrel and eight placebo patients required TLR (P = 0.04). The clopidogrel patients who needed revascularisation were both resistant to clopidogrel. Minor bleeding complications occurred in one clopidogrel and two placebo patients. CONCLUSION: Dual antiplatetet therapy reduces peri-interventional platelet activation and improves functional outcome without higher bleeding complications. An individual tailored dual antiplatelet therapy seems desirable for endovascularly treated patients with PAD.


Assuntos
Angioplastia/efeitos adversos , Aspirina/uso terapêutico , Doença Arterial Periférica/cirurgia , Inibidores da Agregação Plaquetária/uso terapêutico , Trombose/tratamento farmacológico , Trombose/etiologia , Idoso , Método Duplo-Cego , Feminino , Humanos , Masculino , Doença Arterial Periférica/complicações , Resultado do Tratamento
6.
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
7.
Cardiovasc Intervent Radiol ; 35(6): 1439-47, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22159909

RESUMO

PURPOSE: To evaluate in vivo the role of RAGE (receptor for advanced glycated end products) in the development of restenosis and neointimal proliferation in RAGE-deficient knockout (KO) mice compared with wild-type (WT) mice in an animal model. MATERIALS AND METHODS: Sixteen WT and 15 RAGE-deficient mice underwent microvascular denudation of the common femoral artery under general anaesthesia. Contralateral arteries underwent a sham operation and served as controls. Four weeks after the intervention, all animals were killed, and paraformaldehyde-fixed specimens of the femoral artery were analysed with different stains (hematoxylin and eosin and Elastica van Gieson) and several different types of immunostaining (proliferating cell nuclear antigen, α-actin, collagen, von Willebrand factor, RAGE). Luminal area, area of the neointima, and area of the media were measured in all specimens. In addition, colony-formation assays were performed, and collagen production by WT smooth muscle cells (SMCs) and RAGE-KO SMCs was determined. For statistical analysis, P < 0.05 was considered statistically significant. RESULTS: Four weeks after denudation, WT mice showed a 49.6% loss of luminal area compared with 14.9% loss of luminal area in RAGE-deficient mice (sham = 0% loss) (P < 0.001). The neointima was 18.2 (*1000 µm(2) [n = 15) in the WT group compared with only 8.4 (*1000 µm(2) [n = 16]) in the RAGE-KO group. RAGE-KO SMCs showed significantly decreased proliferation activity and production of extracellular matrix protein. CONCLUSION: RAGE may be shown to play a considerable role in the formation of neointima leading to restenosis after vascular injury.


Assuntos
Artéria Femoral/cirurgia , Neointima , Receptores Imunológicos/metabolismo , Actinas/metabolismo , Animais , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Camundongos , Camundongos Knockout , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro/análise , Receptor para Produtos Finais de Glicação Avançada , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de von Willebrand/metabolismo
8.
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
9.
Invest Radiol ; 46(1): 71-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21139503

RESUMO

RATIONALE AND OBJECTIVES: The objective of this study was to evaluate effects of 6 commercially available magnetic resonance contrast media (CM) on human umbilical vein endothelial cells (HUVEC) and the induction of transforming growth factor-beta (TGF-ß) in dermal fibroblasts (HSF) as a possible model for the pathogenesis of nephrogenic systemic fibrosis. METHODS: HUVECs were incubated with 10× and 20× of the molar standard blood concentration achieved with CM applications for magnetic resonance imaging examinations (10× and 20× concentration) for 24 hours using gadolinium-based CM Gadovist, Magnevist, Multihance, and Omniscan, as well as Teslascan (Manganese-based), and Resovist (Iron-based). Proliferation kinetics (PK), colony formation, and viability assays were performed. Additionally, human dermal fibroblasts (HSF) were incubated for 24 hours with 1× and 20× concentration in all 6 CM, and TGF-ß levels were assessed directly after the incubation period as well as on days 3 and 8 postincubation. RESULTS: HUVEC PK data show similar gains in cell numbers for all 6 CM in both concentration groups over the 17-day assessment period. Only cells incubated with Omniscan and Teslascan differed from the other groups on days 3 and 7 postincubation (P < 0.05). After day 7, a cell regain occurred in the Omniscan and Teslascan groups reaching the numbers of the other groups in sequel. Differences in colony formation were consistent with PK results with a statistically significant reduction in clonogenic activity for Teslascan and Omniscan in HUVEC cells, P < 0.05. No reduction in viability was seen for all groups and conditions. TGF-ß expression of HSF cells incubated with 1× concentration and all CM did not differ significantly from control cells for any point in time investigated. At 20× concentration directly after incubation, TGF-ß was significantly reduced for the Teslascan and Resovist group as 3 compared with control and all other CM groups, P < 0.05. On day 3 postincubation, only Resovist-incubated HSF cells showed a significant reduction of TGF-ß (1.614, standard deviations: 89) as compared with the control group (2.883, standard deviations: 30) and the other CM. TGF-ß was slightly reduced for all CM groups 8 days after incubation (not statistically significant, P > 0.05). CONCLUSIONS: After 24 hours of incubation with Omniscan and Teslascan (10× and 20× concentration), considerable short-term antiproliferative effects in HUVECs were observed. HSF cells (20× concentration) showed a reduction of TGF-ß for Resovist and Teslascan directly after incubation, whereas TGF-ß levels in HSF cells were slightly reduced for all CM 8 days after incubation. Therefore, TGF-ß-mediated proliferative effects on fibroblasts or on collagen synthesis potentially leading to nephrogenic systemic fibrosis may mainly be triggered by tissue monocytes and macrophages in the peripheral blood instead of dermal fibroblasts.


Assuntos
Meios de Contraste/efeitos adversos , Endotélio Vascular/citologia , Imageamento por Ressonância Magnética/métodos , Dermopatia Fibrosante Nefrogênica/diagnóstico , Fator de Crescimento Transformador beta1/biossíntese , Veias Umbilicais/citologia , Colágeno , Fibroblastos/efeitos dos fármacos , Gadolínio DTPA/efeitos adversos , Humanos , Imunoensaio , Imageamento por Ressonância Magnética/instrumentação , Dermopatia Fibrosante Nefrogênica/diagnóstico por imagem , Dermopatia Fibrosante Nefrogênica/patologia , Cintilografia , Fatores de Tempo
10.
Cardiovasc Intervent Radiol ; 34(4): 816-23, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21046387

RESUMO

PURPOSE: Rhenium-186 ((186)Re) and rhenium-188 ((188)Re) are promising radionuclides for the inhibition of restenosis after percutaneous transluminal angioplasty or other vascular interventions. Until now the maximal dose tolerance of endothelial cells has not been clearly known. MATERIALS AND METHODS: To characterize the effects of local irradiation treatment, human aortic endothelial cells (ECs) were incubated with different doses of (186)Re and (188)Re. Two days after plating, ECs received treatment for a period of 5 days. The total radiation doses applied were 1, 4, 8, 16, and 32 Gy. On days 1, 3, 5, 7, and 12 after initial rhenium incubation, cell growth, clonogenic activity, cell-cycle distribution, and cytoskeletal architecture were evaluated. RESULTS: From the first day on, a dose-dependent growth inhibition was observed. Cumulative doses of ≥32 Gy caused a weak colony formation and significant alterations in the cytoskeletal architecture. An increased fraction of cells in G2/M phase was seen for cumulative radiation doses of ≥16 Gy. Interestingly, there were no significant differences between (186)Re and (188)Re. CONCLUSION: Even for low dose rates of ß particles a dose-dependent proliferation inhibition of ECs is seen. Doses beyond 32 Gy alter the cytoskeletal architecture with possibly endothelial dysfunction and late thrombosis.


Assuntos
Aorta/citologia , Proliferação de Células/efeitos da radiação , Radioisótopos/farmacologia , Rênio/farmacologia , Angioplastia , Ciclo Celular/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias , Relação Dose-Resposta à Radiação , Células Endoteliais , Técnicas In Vitro , Dose Máxima Tolerável , Microscopia de Fluorescência
11.
Invest Radiol ; 45(9): 513-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20661142

RESUMO

RATIONALE AND OBJECTIVES: The objective of this investigation was to evaluate 6 magnetic resonance contrast media (CM) with regard to their different effects on human embryonic lung fibroblasts (HEL-299). METHODS: Human embryonic fibroblasts (HEL-299) were incubated with 1x, 5x, 10x, and 20x of the normal molar blood concentration (1x, 5x, 10x, 20x conc.) reached through routine contrast media applications for MRI examinations. Four gadolinium-based CM, ie, Gadovist, Magnevist, Multihance, Omniscan, Teslascan (Manganese-based), and Resovist (Iron-based), with incubation periods over 4 hours and 24 hours were investigated. Proliferation kinetics, colony formation, and viability assays were performed after 4 and 24 hours of treatment. Apoptotic cells were quantified after tetramethylrhodamine ethyl ester staining following 24 hours of CM media incubation (20x conc.) by fluorescence activated cell sorting cytometry. Furthermore, immunofluorescence images with vimentin staining were obtained (20x conc., 24 hours treatment). Cell cycle analysis was performed after 24 hours of incubation and 20x conc. directly after incubation and 24 hours later (fluorescence activated cell sorting cytometry). RESULTS: The proliferation kinetics performed with 20x conc. revealed a persistent increase in cell numbers until day 11 for all CM without significant differences after 4 hours of incubation. A significant reduction in initial cell numbers was recorded in the 24-hours-group after 4 days of CM incubation with Magnevist, Multihance, Omniscan, and Teslascan. Solely cells incubated with Resovist and Gadovist failed to show decreased cell numbers when compared with the control group. However, a considerable cell regain occurred afterward reaching control-group levels on day 21. Colony numbers were significantly reduced (about 20%, respectively) with Magnevist at 10x and 20x conc., as well as Omniscan and Multihance at 20x conc. when compared with all other groups, P < 0.05. Cell-cycle distribution showed a reduction of S-phase cells for Magnevist, Omniscan, and Multihance (2.9%-10.5%) when compared with Gadovist, Resovist and Teslascan (16.7%-21.0%). Twenty-four hours after incubation, the percentiles of cells in S-phase were significantly increased for Magnevist, Omniscan, and Multihance (31.4%-38.5%) when compared with Gadovist, Resovist, and Teslascan (18.6%-26.8%), P < 0.05. Viability was not impaired by administration of any CM and no apoptosis was seen after tetramethylrhodamine ethyl ester staining at 24 hours of incubation. Cell morphology remained unchanged in vimentin-staining for all CM and conditioning regimens. CONCLUSIONS: No toxic effects on embryonic fetal lung fibroblasts were detectable after 4 and 24 hours of incubation in 6 MRI CM and 10x to 20x conc. in our setting. Antiproliferative effects, initially detected with Magnevist, Omniscan and Multihance, were rapidly compensated for.


Assuntos
Meios de Contraste , Fibroblastos/citologia , Gadolínio , Pulmão/citologia , Imageamento por Ressonância Magnética/métodos , Análise de Variância , Apoptose , Técnicas de Cultura de Células , Fibroblastos/efeitos dos fármacos , Imunofluorescência , Corantes Fluorescentes , Humanos , Pulmão/efeitos dos fármacos , Imageamento por Ressonância Magnética/instrumentação , Rodaminas
12.
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
13.
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
14.
Cardiovasc Intervent Radiol ; 32(4): 737-44, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19396490

RESUMO

The purpose of this study was to compare the beneficial effects of radioactive stents and radioactive stents plus additional chemotherapy in the palliative treatment of cholangiocellular carcinomas. Cholangiocellular carcinoma cells (TFK-1 cells) were treated either with 8 Gy (RTB group) or 16 Gy (RTA group) (188)Re or with (188)Re irradiation (8 Gy) combined with either gemcitabine (8 Gy/Gem) or 5-fluorouracil (8 Gy/5-FU) at a dosage of 20 microg/ml medium for 4 days and subsequently compared with an untreated control group. Proliferation kinetics were assessed on days 4, 7, 11, 18, 25, and 32. Colony formation assays were performed on days 7, 18, and 32 and cell cycle distribution was examined on days 4, 7, 11, 15, 25, and 39. Cell proliferation kinetics showed the lowest cell numbers in the 8 Gy/5-FU group (control, 15,390,000; RTA group, 8,394,000; RTB group, 5,609,000; 8 Gy/Gem group, 423,000; and 8 Gy/5-FU group, 297,667). In contrast, clonogenic activity on day 32 was lower in the 8 Gy/Gem group (control, 29.3 colonies; RTB group, 23.1 colonies; 8 Gy/5-FU group, 21.5 colonies; 8 Gy/Gem, 3.3 colonies; and even augmented in the RTA group, with 37.7 colonies). Cell cycle distribution showed similar curves for all groups on slightly different levels except for the 8 Gy/5-FU group, which showed a relatively augmented percentage of cells on day 7 in the G2 M cycle phase and on day 4 in the S phase. In conclusion, irradiation (8 Gy) with (188)Re administered, e.g., via coated stents, combined with Gem could be a valid option for the treatment of CCCs.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/radioterapia , Desoxicitidina/análogos & derivados , Fluoruracila/farmacologia , Radioisótopos/farmacologia , Rênio/farmacologia , Stents , Análise de Variância , Ciclo Celular , Desoxicitidina/farmacologia , Citometria de Fluxo , Humanos , Técnicas In Vitro , Cuidados Paliativos , Células Tumorais Cultivadas , Gencitabina
15.
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
16.
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
17.
J Magn Reson Imaging ; 26(5): 1308-14, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17969164

RESUMO

PURPOSE: To evaluate the effects of the static magnetic field and typical imaging sequences of a high-field magnetic resonance scanner (3 Tesla) on the induction of double-strand breaks (DSBs) in two different human cell lines. MATERIALS AND METHODS: Human promyelocytic leukemia cells (HL-60) and human acute myeloid leukemia cells (KG-1a) were exposed to the static magnetic field alone and to turbo spin-echo (TSE) and gradient-echo (GE) sequences. Flow cytometry was used to quantify gammaH2AX (serine 139 phosphorylated form of histone H2AX) expression of antibody-stained cells as a marker for deoxyribonucleic acid (DNA) DSBs one hour and 24 hours after magnetic field exposure. X-ray-treated cells were used as positive control. RESULTS: Neither exposure to the static magnetic field alone nor to the applied imaging sequences showed significant differences in gammaH2AX expression between exposed and sham-exposed cells. X-ray-treated cells as positive control showed a significant increase in gammaH2AX expression. CONCLUSION: The static magnetic field alone and MRI sequences at 3 Tesla have no effect on the induction of DSBs in HL-60 and KG-1a cells.


Assuntos
Dano ao DNA , DNA de Neoplasias/genética , DNA de Neoplasias/efeitos da radiação , Histonas/genética , Histonas/efeitos da radiação , Imageamento por Ressonância Magnética , Linhagem Celular , Relação Dose-Resposta à Radiação , Células HL-60 , Humanos , Doses de Radiação
18.
J Magn Reson Imaging ; 26(5): 1210-5, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17969170

RESUMO

PURPOSE: To evaluate the influence of magnetic resonance imaging (MRI) on gene expression in embryonic human lung fibroblasts (Hel 299). MATERIALS AND METHODS: The cells were exposed to the static magnetic field and to a turbo spin-echo sequence of an MR scanner at 3.0 Tesla. An MR group (exposed) and a control group (sham-exposed) were set up using a special MR-compatible incubation system. The exposure time was two hours. Gene expression profiles were studied using a complementary deoxyribonucleic acid (cDNA) microarray containing 498 known genes involved in transcription, intracellular transport, structure/junction/adhesion or extracellular matrix, signaling, host defense, energetics, metabolism, cell shape, and death. RESULTS: No changes in gene expression were found in either group (exposed or sham-exposed cells) at the end of a two-hour exposure for any of the 498 tested protein genes. CONCLUSION: The results suggest that MRI has no influence on protein-gene expression in eugenic human lung cells.


Assuntos
Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Expressão Gênica/fisiologia , Expressão Gênica/efeitos da radiação , Pulmão/citologia , Pulmão/metabolismo , Imageamento por Ressonância Magnética , Proteínas/metabolismo , Linhagem Celular , Relação Dose-Resposta à Radiação , Campos Eletromagnéticos , Fibroblastos/citologia , Humanos , Pulmão/embriologia , Doses de Radiação
19.
Radiology ; 244(2): 514-23, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17562811

RESUMO

PURPOSE: To prospectively evaluate the influence of superparamagnetic iron oxide (SPIO) or ultrasmall SPIO (USPIO) particles on the surface epitope pattern of adult mesenchymal stem cells (MSCs) by regulating the expression of transferrin receptor and to prospectively evaluate the influence of transfection agents (TAs) on the uptake of SPIO or USPIO particles in MSCs. MATERIALS AND METHODS: The study was approved by the institutional animal care committee of the University of Tübingen. MSCs were isolated from the bone marrow of four rats. To obtain highly homogeneous MSC populations, MSCs from one rat were single-cell cloned. One MSC clone was characterized and selected for the labeling experiments. The MSCs, which were characterized with flow cytometry and in vitro differentiation, were labeled with 200 microg/mL SPIO or USPIO or with 60 microg/mL SPIO or USPIO in combination with TAs. Aggregations of labeled cells were accommodated inside a defined volume in an agar gel matrix. Magnetic resonance (MR) imaging was performed to measure SPIO- or USPIO-induced signal voids. Quantification of cellular total iron load (TIL) (intracellular iron plus iron coating the cellular surface), determination of cellular viability, and electron microscopy were also performed. RESULTS: Labeling of MSCs with SPIO or USPIO was feasible without affecting cell viability (91.1%-94.7%) or differentiation potential. For MR imaging, SPIO plus a TA was most effective, depicting 5000 cells with an average TIL of 76.5 pg per cell. SPIO or USPIO particles in combination with TAs coated the cellular surface but were not incorporated into cells. In nontransfected cells, SPIO or USPIO was taken up. MSCs labeled with SPIO or USPIO but without a TA showed enhanced expression of transferrin receptor, in contrary to both MSCs labeled with SPIO or USPIO and a TA and control cells. CONCLUSION: SPIO or USPIO labeling without TAs has an influence on gene expression of MSCs upregulating transferrin receptor. Furthermore, SPIO labeling with a TA will coat the cellular surface.


Assuntos
Meios de Contraste/farmacocinética , Ferro/farmacocinética , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais , Óxidos/farmacocinética , Receptores da Transferrina/metabolismo , Animais , Dextranos , Estudos de Viabilidade , Óxido Ferroso-Férrico , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Nanopartículas de Magnetita , Masculino , Tamanho da Partícula , Estudos Prospectivos , Ratos , Ratos Sprague-Dawley , Transfecção , Regulação para Cima
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA