Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 62017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28695825

RESUMO

Increased mTORC1 signaling from TSC1/TSC2 inactivation is found in cancer and causes tuberous sclerosis complex (TSC). The role of mesenchymal-derived cells in TSC tumorigenesis was investigated through disruption of Tsc2 in craniofacial and limb bud mesenchymal progenitors. Tsc2cKOPrrx1-cre mice had shortened lifespans and extensive hamartomas containing abnormal tortuous, dilated vessels prominent in the forelimbs. Abnormalities were blocked by the mTORC1 inhibitor sirolimus. A Tsc2/mTORC1 expression signature identified in Tsc2-deficient fibroblasts was also increased in bladder cancers with TSC1/TSC2 mutations in the TCGA database. Signature component Lgals3 encoding galectin-3 was increased in Tsc2-deficient cells and serum of Tsc2cKOPrrx1-cre mice. Galectin-3 was increased in TSC-related skin tumors, angiomyolipomas, and lymphangioleiomyomatosis with serum levels in patients with lymphangioleiomyomatosis correlating with impaired lung function and angiomyolipoma presence. Our results demonstrate Tsc2-deficient mesenchymal progenitors cause aberrant morphogenic signals, and identify an expression signature including Lgals3 relevant for human disease of TSC1/TSC2 inactivation and mTORC1 hyperactivity.


Assuntos
Galectina 3/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Células-Tronco Mesenquimais/fisiologia , Neoplasias Cutâneas/fisiopatologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Sanguíneas , Galectinas , Humanos , Camundongos , Camundongos Knockout , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/deficiência
2.
Sci Rep ; 7(1): 2093, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28522833

RESUMO

The regulatory control of cardiac endoplasmic reticulum (ER) stress is incompletely characterized. As ER stress signaling upregulates the E3-ubiquitin ligase Parkin, we investigated the role of Parkin in cardiac ER stress. Parkin knockout mice exposed to aortic constriction-induced cardiac pressure-overload or in response to systemic tunicamycin (TM) developed adverse ventricular remodeling with excessive levels of the ER regulatory C/EBP homologous protein CHOP. CHOP was identified as a Parkin substrate and its turnover was Parkin-dose and proteasome-dependent. Parkin depletion in cardiac HL-1 cells increased CHOP levels and enhanced susceptibility to TM-induced cell death. Parkin reconstitution rescued this phenotype and the contribution of excess CHOP to this ER stress injury was confirmed by reduction in TM-induced cell death when CHOP was depleted in Parkin knockdown cardiomyocytes. Isogenic Parkin mutant iPSC-derived cardiomyocytes showed exaggerated ER stress induced CHOP and apoptotic signatures and myocardium from subjects with dilated cardiomyopathy showed excessive Parkin and CHOP induction. This study identifies that Parkin functions to blunt excessive CHOP to prevent maladaptive ER stress-induced cell death and adverse cardiac ventricular remodeling. Additionally, Parkin is identified as a novel post-translational regulatory moderator of CHOP stability and uncovers an additional stress-modifying function of this E3-ubiquitin ligase.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Estresse do Retículo Endoplasmático , Miócitos Cardíacos/metabolismo , Fator de Transcrição CHOP/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Cardiomiopatia Dilatada/patologia , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Ubiquitina-Proteína Ligases/genética , Remodelação Ventricular
3.
Cardiovasc Revasc Med ; 17(2): 106-12, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26874740

RESUMO

INTRODUCTION: Nanoparticles may serve as a promising means to deliver novel therapeutics to the myocardium following myocardial infarction. We sought to determine whether lipid-based liposomal nanoparticles can be shown through different imaging modalities to specifically target injured myocardium following intravenous injection in an ischemia-reperfusion murine myocardial infarction model. METHODS: Mice underwent ischemia-reperfusion surgery and then either received tail-vein injection with gadolinium- and fluorescent-labeled liposomes or no injection (control). The hearts were harvested 24h later and underwent T1 and T2-weighted ex vivo imaging using a 7 Tesla Bruker magnet. The hearts were then sectioned for immunohistochemistry and optical fluorescent imaging. RESULTS: The mean size of the liposomes was 100nm. T1-weighted signal intensity was significantly increased in the ischemic vs. the non-ischemic myocardium for mice that received liposomes compared with control. Optical imaging demonstrated significant fluorescence within the infarct area for the liposome group compared with control (163±31% vs. 13±14%, p=0.001) and fluorescent microscopy confirmed the presence of liposomes within the ischemic myocardium. CONCLUSIONS: Liposomes traffic to the heart and preferentially home to regions of myocardial injury, enabling improved diagnosis of myocardial injury and could serve as a vehicle for drug delivery.


Assuntos
Albuminas/farmacocinética , Meios de Contraste/farmacocinética , Corantes Fluorescentes/farmacocinética , Gadolínio DTPA/farmacocinética , Imageamento por Ressonância Magnética , Infarto do Miocárdio/diagnóstico por imagem , Traumatismo por Reperfusão Miocárdica/diagnóstico por imagem , Miocárdio/metabolismo , Imagem Óptica/métodos , Fosfatidiletanolaminas/farmacocinética , Albuminas/administração & dosagem , Animais , Meios de Contraste/administração & dosagem , Modelos Animais de Doenças , Corantes Fluorescentes/administração & dosagem , Gadolínio DTPA/administração & dosagem , Imuno-Histoquímica , Injeções Intravenosas , Lipossomos , Masculino , Camundongos , Microscopia de Fluorescência , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Nanopartículas , Tamanho da Partícula , Fosfatidiletanolaminas/administração & dosagem , Distribuição Tecidual
4.
Cardiovasc Res ; 106(3): 478-87, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25858253

RESUMO

AIMS: Atypical chemokine receptor 1 (Ackr1; previously known as the Duffy antigen receptor for chemokines or Darc) is thought to regulate acute inflammatory responses in part by scavenging inflammatory CC and CXC chemokines; however, evidence for a role in chronic inflammation has been lacking. Here we investigated the role of Ackr1 in chronic inflammation, in particular in the setting of atherogenesis, using the apolipoprotein E-deficient (ApoE(-/-)) mouse model. METHODS AND RESULTS: Ackr1(-/-)ApoE(-/-) and Ackr1(+/+)ApoE(-/-) littermates were obtained by crossing ApoE(-/-) mice and Ackr1(-/-) mice on a C57BL/6J background. Ackr1 (+/+)ApoE(-/-)mice fed a Western diet up-regulated Ackr1 expression in the aorta and had markedly increased atherosclerotic lesion size compared with Ackr1(-/-)ApoE(-/-) mice. This difference was observed in both the whole aorta and the aortic root in both early and late stages of the model. Ackr1 deficiency did not affect serum cholesterol levels or macrophage, collagen or smooth muscle cell content in atherosclerotic plaques, but significantly reduced the expression of Ccl2 and Cxcl1 in the whole aorta of ApoE(-/-) mice. In addition, Ackr1 deficiency resulted in a modest decrease in T cell subset frequency and inflammatory mononuclear phagocyte content in aorta and blood in the model. CONCLUSIONS: Ackr1 deficiency appears to be protective in the ApoE knockout model of atherogenesis, but it is associated with only modest changes in cytokine and chemokine expression as well as T-cell subset frequency and inflammatory macrophage content.


Assuntos
Aorta , Aortite , Apolipoproteínas E , Aterosclerose , Receptores de Superfície Celular , Animais , Feminino , Transferência Adotiva , Aorta/imunologia , Aorta/metabolismo , Aorta/patologia , Aortite/genética , Aortite/imunologia , Aortite/metabolismo , Aortite/patologia , Aortite/prevenção & controle , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Dieta Ocidental , Modelos Animais de Doenças , Sistema do Grupo Sanguíneo Duffy/genética , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Placa Aterosclerótica , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/transplante , Fatores de Tempo
5.
PLoS One ; 10(1): e0115861, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25615642

RESUMO

Congenital heart valve defects in humans occur in approximately 2% of live births and are a major source of compromised cardiac function. In this study we demonstrate that normal heart valve development and cardiac function are dependent upon Galnt1, the gene that encodes a member of the family of glycosyltransferases (GalNAc-Ts) responsible for the initiation of mucin-type O-glycosylation. In the adult mouse, compromised cardiac function that mimics human congenital heart disease, including aortic and pulmonary valve stenosis and regurgitation; altered ejection fraction; and cardiac dilation, was observed in Galnt1 null animals. The underlying phenotype is aberrant valve formation caused by increased cell proliferation within the outflow tract cushion of developing hearts, which is first detected at developmental stage E11.5. Developing valves from Galnt1 deficient animals displayed reduced levels of the proteases ADAMTS1 and ADAMTS5, decreased cleavage of the proteoglycan versican and increased levels of other extracellular matrix proteins. We also observed increased BMP and MAPK signaling. Taken together, the ablation of Galnt1 appears to disrupt the formation/remodeling of the extracellular matrix and alters conserved signaling pathways that regulate cell proliferation. Our study provides insight into the role of this conserved protein modification in cardiac valve development and may represent a new model for idiopathic valve disease.


Assuntos
Embrião de Mamíferos/fisiopatologia , Cardiopatias Congênitas/patologia , Valvas Cardíacas/fisiopatologia , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAMTS1 , Proteína ADAMTS5 , Animais , Proliferação de Células , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/fisiopatologia , Valvas Cardíacas/patologia , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Polipeptídeo N-Acetilgalactosaminiltransferase
6.
Nat Rev Cardiol ; 7(2): 97-105, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20027188

RESUMO

Despite promising preclinical data, the treatment of cardiovascular diseases using embryonic, bone-marrow-derived, and skeletal myoblast stem cells has not yet come to fruition within mainstream clinical practice. Major obstacles in cardiac stem cell investigations include the ability to monitor cell engraftment and survival following implantation within the myocardium. Several cellular imaging modalities, including reporter gene and MRI-based tracking approaches, have emerged that provide the means to identify, localize, and monitor stem cells longitudinally in vivo following implantation. This Review will examine the various cardiac cellular tracking modalities, including the combinatorial use of several probes in multimodality imaging, with a focus on data from the past 5 years.


Assuntos
Diagnóstico por Imagem , Cardiopatias/diagnóstico , Cardiopatias/cirurgia , Miocárdio/patologia , Regeneração , Transplante de Células-Tronco , Animais , Meios de Contraste , Diagnóstico por Imagem/métodos , Óxido Ferroso-Férrico , Gadolínio , Genes Reporter , Cardiopatias/patologia , Humanos , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Valor Preditivo dos Testes , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Resultado do Tratamento
7.
Circ Res ; 105(7): 705-12, 11 p following 712, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19696408

RESUMO

RATIONALE: Exercise capacity is a physiological characteristic associated with protection from both cardiovascular and all-cause mortality. p53 regulates mitochondrial function and its deletion markedly diminishes exercise capacity, but the underlying genetic mechanism orchestrating this is unclear. Understanding the biology of how p53 improves exercise capacity may provide useful insights for improving both cardiovascular as well as general health. OBJECTIVE: The purpose of this study was to understand the genetic mechanism by which p53 regulates aerobic exercise capacity. METHODS AND RESULTS: Using a variety of physiological, metabolic, and molecular techniques, we further characterized maximum exercise capacity and the effects of training, measured various nonmitochondrial and mitochondrial determinants of exercise capacity, and examined putative regulators of mitochondrial biogenesis. As p53 did not affect baseline cardiac function or inotropic reserve, we focused on the involvement of skeletal muscle and now report a wider role for p53 in modulating skeletal muscle mitochondrial function. p53 interacts with Mitochondrial Transcription Factor A (TFAM), a nuclear-encoded gene important for mitochondrial DNA (mtDNA) transcription and maintenance, and regulates mtDNA content. The increased mtDNA in p53(+/+) compared to p53(-/-) mice was more marked in aerobic versus glycolytic skeletal muscle groups with no significant changes in cardiac tissue. These in vivo observations were further supported by in vitro studies showing overexpression of p53 in mouse myoblasts increases both TFAM and mtDNA levels whereas depletion of TFAM by shRNA decreases mtDNA content. CONCLUSIONS: Our current findings indicate that p53 promotes aerobic metabolism and exercise capacity by using different mitochondrial genes and mechanisms in a tissue-specific manner.


Assuntos
DNA Mitocondrial/metabolismo , Tolerância ao Exercício , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Esforço Físico , Proteína Supressora de Tumor p53/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Tolerância ao Exercício/genética , Glicólise/genética , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Força Muscular , Mutação , Miocárdio/metabolismo , Consumo de Oxigênio , Interferência de RNA , Elementos de Resposta , Natação , Fatores de Tempo , Transdução Genética , Transfecção , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Regulação para Cima , Função Ventricular Esquerda
8.
FASEB J ; 22(9): 3234-46, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18556461

RESUMO

This study investigated the factors responsible for migration and homing of magnetically labeled AC133(+) cells at the sites of active angiogenesis in tumor. AC133(+) cells labeled with ferumoxide-protamine sulfate were mixed with either rat glioma or human melanoma cells and implanted in flank of nude mice. An MRI of the tumors including surrounding tissues was performed. Tumor sections were stained for Prussian blue (PB), platelet-derived growth factor (PDGF), hypoxia-inducible factor-1alpha (HIF-1alpha), stromal cell derived factor-1 (SDF-1), matrix metalloproteinase-2 (MMP-2), vascular endothelial growth factor (VEGF), and endothelial markers. Fresh snap-frozen strips from the central and peripheral parts of the tumor were collected for Western blotting. MRIs demonstrated hypointense regions at the periphery of the tumors where the PB(+)/AC133(+) cells were positive for endothelial cells markers. At the sites of PB(+)/AC133(+) cells, both HIF-1alpha and SDF-1 were strongly positive and PDGF and MMP-2 showed generalized expression in the tumor and surrounding tissues. There was no significant association of PB(+)/AC133(+) cell localization and VEGF expression in tumor cells. Western blot demonstrated strong expression of the SDF-1, MMP-2, and PDGF at the peripheral parts of the tumors. HIF-1alpha was expressed at both the periphery and central parts of the tumor. This work demonstrates that magnetically labeled cells can be used as probes for MRI and histological identification of administered cells.


Assuntos
Movimento Celular , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Mesenquimais , Neovascularização Patológica/patologia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Quimiocina CXCL12/biossíntese , Dextranos , Feminino , Óxido Ferroso-Férrico , Glioma/patologia , Glicoproteínas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Ferro , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita , Metaloproteinase 2 da Matriz/biossíntese , Melanoma Amelanótico/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Óxidos , Peptídeos/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , Protaminas , Ratos , Fator A de Crescimento do Endotélio Vascular/biossíntese
9.
Mol Imaging ; 6(3): 212-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17532887

RESUMO

Superparamagnetic iron oxide (SPIO) nanoparticles, either modified or in combination with other macromolecules, are being used for magnetic labeling of stem cells and other cells to monitor cell trafficking by magnetic resonance imaging (MRI) in experimental models. The correlation of histology to MRI depends on the ability to detect SPIO-labeled cells using Prussian blue (PB) stain and fluorescent tags to cell surface markers. Exposure of PB-positive sections to ultraviolet light at a wavelength of 365 nm commonly used fluorescence microscopy can result in color transformation of PB-positive material from blue to brown. Although the PB color transformation is primarily an artifact that may occur during fluorescence microscopy, the transformation can be manipulated using imaging process software for the detection of low levels of iron labeled cells in tissues samples.


Assuntos
Artefatos , Cor , Corantes/química , Ferrocianetos/química , Óxido Ferroso-Férrico/química , Nanopartículas Metálicas/química , Microscopia de Fluorescência , Coloração e Rotulagem/métodos , Animais , Humanos , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/citologia , Ratos , Raios Ultravioleta
10.
NMR Biomed ; 20(3): 200-15, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17451184

RESUMO

MRI has contributed to significant advances in the understanding of neurological diseases in humans. It has also been used to evaluate the spectrum of mouse models spanning from developmental abnormalities during embryogenesis, evaluation of transgenic and knockout models, through various neurological diseases such as stroke, tumors, degenerative and inflammatory diseases. The MRI techniques used clinically are technically more challenging in the mouse because of the size of the brain; however, mouse imaging provides researchers with the ability to explore cellular and molecular imaging that one day may translate into clinical practice. This article presents an overview of the use of MRI in mouse models of a variety of neurological disorders and a brief review of cellular imaging using magnetically tagged cells in the mouse central nervous system.


Assuntos
Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Doenças do Sistema Nervoso/diagnóstico , Animais , Humanos , Camundongos
11.
J Exp Med ; 203(5): 1235-47, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16636132

RESUMO

Emerging evidence suggests that both human stem cells and mature stromal cells can play an important role in the development and growth of human malignancies. In contrast to these tumor-promoting properties, we observed that in an in vivo model of Kaposi's sarcoma (KS), intravenously (i.v.) injected human mesenchymal stem cells (MSCs) home to sites of tumorigenesis and potently inhibit tumor growth. We further show that human MSCs can inhibit the in vitro activation of the Akt protein kinase within some but not all tumor and primary cell lines. The inhibition of Akt activity requires the MSCs to make direct cell-cell contact and can be inhibited by a neutralizing antibody against E-cadherin. We further demonstrate that in vivo, Akt activation within KS cells is potently down-regulated in areas adjacent to MSC infiltration. Finally, the in vivo tumor-suppressive effects of MSCs correlates with their ability to inhibit target cell Akt activity, and KS tumors engineered to express a constitutively activated Akt construct are no longer sensitive to i.v. MSC administration. These results suggest that in contrast to other stem cells or normal stromal cells, MSCs possess intrinsic antineoplastic properties and that this stem cell population might be of particular utility for treating those human malignancies characterized by dysregulated Akt.


Assuntos
Efeito Enxerto vs Tumor/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Sarcoma de Kaposi/imunologia , Animais , Modelos Animais de Doenças , Ativação Enzimática/imunologia , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Proteína Oncogênica v-akt/imunologia , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/terapia , Células Estromais/imunologia , Células Estromais/transplante , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Invest Radiol ; 41(3): 332-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16481917

RESUMO

OBJECTIVES: The objectives of this study were to test cell-labeling methods to achieve intracellular labeling and T1 enhancement of cells on magnetic resonance imaging using a paramagnetic Gd@C82 fullerenol contrast agent, and to determine the effect of labeling on cell viability, metabolism, and differentiation capacity. MATERIALS AND METHODS: We tested the use of a transfection agent for labeling cells in culture with Gd@C82 fullerenol. Proliferation, viability, and differentiation assays of mesenchymal stem cell (MSC) cultures; light and electron microscopy of MSC and macrophages; and MRI of MSC, macrophage, and HeLa cervical carcinoma cell cultures in vitro and in vivo were performed to evaluate the labeled cells. RESULTS: Protamine sulfate transfection increased cell uptake of Gd@C82 fullerenols. The label was distributed in endosomes in the cytoplasm as shown by electron microscopy. High viability was shown for all cell lines and normal differentiation capacity was shown for MSCs. T1 of labeled MSC at 7 T was reduced 71% compared with unlabeled cells. CONCLUSIONS: Cellular labeling with Gd@C82 is feasible and can produce T1-enhanced cells on magnetic resonance imaging. This study suggests that further investigation of Gd fullerenols for tracking studies of viable cells, including stem cells, is warranted.


Assuntos
Meios de Contraste/metabolismo , Fulerenos/metabolismo , Gadolínio DTPA/metabolismo , Células HeLa/metabolismo , Imageamento por Ressonância Magnética , Células-Tronco Mesenquimais/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Estudos de Viabilidade , Células HeLa/citologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica , Ratos , Ratos Sprague-Dawley
13.
Stem Cells ; 24(3): 671-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16179427

RESUMO

UNLABELLED: AC133 cells, a subpopulation of CD34+ hematopoietic stem cells, can transform into endothelial cells that may integrate into the neovasculature of tumors or ischemic tissue. Most current imaging modalities do not allow monitoring of early migration and incorporation of endothelial progenitor cells (EPCs) into tumor neovasculature. The goals of this study were to use magnetic resonance imaging (MRI) to track the migration and incorporation of intravenously injected, magnetically labeled EPCs into the blood vessels in a rapidly growing flank tumor model and to determine whether the pattern of EPC incorporation is related to the time of injection or tumor size. MATERIALS AND METHODS: EPCs labeled with ferumoxide-protamine sulfate (FePro) complexes were injected into mice bearing xenografted glioma, and MRI was obtained at different stages of tumor development and size. RESULTS: Migration and incorporation of labeled EPCs into tumor neovasculature were detected as low signal intensity on MRI at the tumor periphery as early as 3 days after EPC administration in preformed tumors. However, low signal intensities were not observed in tumors implanted at the time of EPC administration until tumor size reached 1 cm at 12 to 14 days. Prussian blue staining showed iron-positive cells at the sites corresponding to low signal intensity on MRI. Confocal microscopy showed incorporation into the neovasculature, and immunohistochemistry clearly demonstrated the transformation of the administered EPCs into endothelial cells. CONCLUSION: MRI demonstrated the incorporation of FePro-labeled human CD34+/AC133+ EPCs into the neovasculature of implanted flank tumors.


Assuntos
Movimento Celular , Glioma/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Angiografia por Ressonância Magnética , Neovascularização Patológica/metabolismo , Antígeno AC133 , Animais , Antígenos CD , Antígenos CD34 , Linhagem Celular , Meios de Contraste/farmacologia , Dextranos , Feminino , Óxido Ferroso-Férrico , Glioma/patologia , Glicoproteínas , Células-Tronco Hematopoéticas/patologia , Humanos , Ferro/farmacologia , Angiografia por Ressonância Magnética/métodos , Nanopartículas de Magnetita , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Confocal/métodos , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neovascularização Patológica/patologia , Óxidos/farmacologia , Peptídeos
14.
Blood ; 105(1): 420-5, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15331444

RESUMO

Bone marrow-derived endothelial precursor cells incorporate into neovasculature and have been successfully used as vehicles for gene delivery to brain tumors. To determine whether systemically administered Sca1+ bone marrow cells labeled with superparamagnetic iron oxide nanoparticles can be detected by in vivo magnetic resonance imaging in a mouse brain tumor model, mouse Sca1+ cells were labeled in vitro with ferumoxides-poly-L-lysine complexes. Labeled or control cells were administered intravenously to glioma-bearing severe combined immunodeficient (SCID) mice. Magnetic resonance imaging (MRI) was performed during tumor growth. Mice that received labeled cells demonstrated hypointense regions within the tumor that evolved over time and developed a continuous dark hypointense ring at a consistent time point. This effect was not cleared by administration of a gadolinium contrast agent. Histology showed iron-labeled cells around the tumor rim in labeled mice, which expressed CD31 and von Willebrand factor, indicating the transplanted cells detected in the tumor have differentiated into endothelial-like cells. These results demonstrate that MRI can detect the incorporation of magnetically labeled bone marrow-derived precursor cells into tumor vasculature as part of ongoing angiogenesis and neovascularization. This technique can be used to directly identify neovasculature in vivo and to facilitate gene therapy by noninvasively monitoring these cells as gene delivery vectors.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Modelos Animais de Doenças , Glioma/irrigação sanguínea , Imageamento por Ressonância Magnética , Células-Tronco/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/patologia , Magnetismo , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Ratos , Transplante de Células-Tronco
15.
Blood ; 104(4): 1217-23, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15100158

RESUMO

Recently, there have been several reports using various superparamagnetic iron oxide (SPIO) nanoparticles to label mammalian cells for monitoring their temporal and spatial migration in vivo by magnetic resonance imaging (MRI). The purpose of this study was to evaluate the efficiency and toxicity of labeling cells using 2 commercially available Food and Drug Administration (FDA)-approved agents, ferumoxides, a suspension of dextran-coated SPIO used as an MRI contrast agent, and protamine sulfate, conventionally used to reverse heparin anticoagulation but also used ex vivo as a cationic transfection agent. After labeling of human mesenchymal stem cells (MSCs) and hematopoietic (CD34+) stem cells and other mammalian cells with ferumoxides-protamine sulfate complexes (FE-Pro), cellular toxicity, functional capacity, and quantitative cellular iron incorporation were determined. FE-Pro-labeled cells demonstrated no short- or long-term toxicity, changes in differentiation capacity of the stem cells, or changes in phenotype when compared with unlabeled cells. Efficient labeling with FE-Pro was observed with iron content per cell varying between 2.01 +/- 0.1 pg for CD34+ cells and 10.94 +/- 1.86 pg for MSCs with 100% of cells labeled. Cell labeling using these agents should facilitate the translation of this method to clinical trials for evaluation of trafficking of infused or transplanted cells by MRI.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Ferro , Células-Tronco Mesenquimais/efeitos dos fármacos , Técnicas de Sonda Molecular , Neoplasias/patologia , Óxidos , Animais , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dextranos , Avaliação de Medicamentos , Óxido Ferroso-Férrico , Células-Tronco Hematopoéticas/citologia , Humanos , Ferro/análise , Ferro/farmacocinética , Ferro/toxicidade , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Células-Tronco Mesenquimais/citologia , Camundongos , Sondas Moleculares/farmacocinética , Sondas Moleculares/toxicidade , Óxidos/farmacocinética , Óxidos/toxicidade , Protaminas/farmacocinética , Protaminas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA