Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Oncoimmunology ; 9(1): 1776577, 2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32923139

RESUMO

Currently, human glioma tumors are mostly modeled in immunodeficient recipients; however, lack of interactions with adaptive immune system is a serious flaw, particularly in the era when immunotherapies dominate treatment strategies. Our group was the first to successfully establish the orthotopic transplantation of human glioblastoma (GBM) in immunocompetent mice by inducing immunological tolerance using a short-term, systemic costimulation blockade strategy (CTLA-4-Ig and MR1). In this study, we further validated the feasibility of this method by modeling pediatric diffuse intrinsic pontine glioma (DIPG) and two types of adult GBM (GBM1, GBM551), in mice with intact immune systems and immunodeficient mice. We found that all three glioma models were successfully established, with distinct difference in tumor growth patterns and morphologies, after orthotopic xenotransplantation in tolerance-induced immunocompetent mice. Long-lasting tolerance that is maintained for up to nearly 200 d in GBM551 confirmed the robustness of this model. Moreover, we found that tumors in immunocompetent mice displayed features more similar to the clinical pathophysiology found in glioma patients, characterized by inflammatory infiltration and strong neovascularization, as compared with tumors in immunodeficient mice. In summary, we have validated the robustness of the costimulatory blockade strategy for tumor modeling and successfully established three human glioma models including the pediatric DIPG whose preclinical study is particularly thwarted by the lack of proper animal models.


Assuntos
Neoplasias do Tronco Encefálico , Glioblastoma , Glioma , Adulto , Animais , Criança , Humanos , Camundongos , Neovascularização Patológica
2.
J Vasc Interv Radiol ; 31(9): 1483-1491, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32800664

RESUMO

PURPOSE: To examine safety and efficacy of bariatric arterial embolization (BAE) with x-ray-visible embolic microspheres (XEMs) and an antireflux catheter in swine. MATERIAL AND METHODS: BAE with selective infusion of XEMs (n = 6) or saline (n = 4, control) into gastric fundal arteries was performed under x-ray guidance. Weight and plasma hormone levels were measured at baseline and weekly for 4 weeks after embolization. Cone-beam CT images were acquired immediately after embolization and weekly for 4 weeks. Hormone-expressing cells in the stomach were assessed by immunohistochemical staining. RESULTS: BAE pigs lost weight 1 week after embolization followed by significantly impaired weight gain relative to control animals (14.3% vs 20.9% at 4 weeks, P = .03). Plasma ghrelin levels were significantly lower in BAE pigs than in control animals (1,221.6 pg/mL vs 1,706.2 pg/mL at 4 weeks, P < .01). XEMs were visible on x-ray and cone-beam CT during embolization, and radiopacity persisted over 4 weeks (165.5 HU at week 1 vs 158.5 HU at week 4, P = .9). Superficial mucosal ulcerations were noted in 1 of 6 BAE animals. Ghrelin-expressing cell counts were significantly lower in the gastric fundus (17.7 vs 36.8, P < .00001) and antrum (24.2 vs 46.3, P < .0001) of BAE pigs compared with control animals. Gastrin-expressing cell counts were markedly reduced in BAE pigs relative to control animals (98.5 vs 127.0, P < .02). Trichrome staining demonstrated significantly more fibrosis in BAE animals compared with control animals (13.8% vs 8.7%, P < .0001). CONCLUSIONS: XEMs enabled direct visualization of embolic material during and after embolization. BAE with XEMs and antireflux microcatheters was safe and effective.


Assuntos
Regulação do Apetite , Comportamento Animal , Catéteres , Embolização Terapêutica/instrumentação , Artéria Gástrica , Fundo Gástrico/irrigação sanguínea , Grelina/sangue , Redução de Peso , Animais , Tomografia Computadorizada de Feixe Cônico , Artéria Gástrica/diagnóstico por imagem , Fundo Gástrico/metabolismo , Fundo Gástrico/patologia , Infusões Intra-Arteriais , Microesferas , Sus scrofa , Fatores de Tempo
3.
Stem Cells Int ; 2019: 9732319, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001343

RESUMO

Stem cell therapies, although promising for treating peripheral arterial disease (PAD), often suffer from low engraftment rates and the inability to confirm the delivery success and track cell distribution and engraftment. Stem cell microencapsulation combined with imaging contrast agents may provide a means to simultaneously enhance cell survival and enable cell tracking with noninvasive imaging. Here, we have evaluated a novel MRI- and X-ray-visible microcapsule formulation for allogeneic mesenchymal stem cell (MSC) delivery and tracking in a large animal model. Bone marrow-derived MSCs from male New Zealand White rabbits were encapsulated using a modified cell encapsulation method to incorporate a dual-modality imaging contrast agent, perfluorooctyl bromide (PFOB). PFOB microcapsules (PFOBCaps) were then transplanted into the medial thigh of normal or PAD female rabbits. In vitro MSC viability remained high (79 ± 5% at 4 weeks of postencapsulation), and as few as two and ten PFOBCaps could be detected in phantoms using clinical C-arm CT and 19F MRI, respectively. Successful injections of PFOBCaps in the medial thigh of normal (n = 15) and PAD (n = 16) rabbits were demonstrated on C-arm CT at 1-14 days of postinjection. Using 19F MRI, transplanted PFOBCaps were clearly identified as "hot spots" and showed one-to-one correspondence to the radiopacities on C-arm CT. Concordance of 19F MRI and C-arm CT locations of PFOBCaps with postmortem locations was high (95%). Immunohistological analysis revealed high MSC survival in PFOBCaps (>56%) two weeks after transplantation while naked MSCs were no longer viable beyond three days after delivery. These findings demonstrate that PFOBCaps could maintain cell viability even in the ischemic tissue and provide a means to monitor cell delivery and track engraftment using clinical noninvasive imaging systems.

4.
PLoS One ; 13(10): e0204650, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30273376

RESUMO

Demyelinating disorders such as multiple sclerosis (MS) or transverse myelitis are devastating neurological conditions with no effective cure. Prevention of myelin loss or restoration of myelin are key for successful therapy. To investigate the disease and develop cures animal models with good clinical relevance are essential. The goal of the current study was to establish a model of focal demyelination in the brain of domestic pig using MRI-guided gliotoxin delivery. The rationale for developing a new myelin disease model in the domestic pig was based on the fact that the brain in pigs is anatomically and histologically much more similar to that of humans compared to the rodent brain. For MRI-assisted gliotoxin injection, eight 30 kg pigs were subjected to treatment with lysolecithin (20, 30 mg/ml); or with ethidium bromide (0.0125, 0.05, 0.2 mg/ml). Animals were placed in an MRI scanner for intraparenchymal targeting of gliotoxin into the corona radiata (250 µl over 1h), with real-time monitoring of toxin distribution on T1 scans and monitoring of lesion evolution over seven days using both T1 and T2 scans. After the last MRI, animals were transcardially perfused and brains were processed for histological and immunofluorescent analysis. Gadolinium-enhanced T1 MRI during injection demonstrated biodistribution of the contrast (as a surrogate marker for toxin distribution) and its diffusion through the brain parenchyma. Lesion induction was confirmed on T2-weighted MRI and histopathology, thus enabling the establishment of optimal doses of gliotoxins. To conclude, MRI-guided focal demyelination in swine is accurate and provides real-time confirmation of gliotoxin, thus facilitating placement of focal lesions with high precision. This new model of focal demyelination can be used for further investigation and development of novel therapeutic approaches.


Assuntos
Doenças Desmielinizantes/induzido quimicamente , Gliotoxina/administração & dosagem , Bainha de Mielina/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Convecção , Gadolínio/administração & dosagem , Imageamento por Ressonância Magnética/métodos , Esclerose Múltipla/induzido quimicamente , Malformações do Sistema Nervoso/induzido quimicamente , Suínos , Distribuição Tecidual/efeitos dos fármacos
5.
Radiology ; 289(1): 83-89, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29989526

RESUMO

Purpose To determine whether microsphere size effects ghrelin expression and weight gain after selective bariatric arterial embolization (BAE) in swine. Materials and Methods BAE was performed in 10 swine by using smaller (100-300 µm; n = 5) or larger (300-500 µm; n = 5) calibrated microspheres into gastric arteries. Nine control pigs underwent a sham procedure. Weight and fasting plasma ghrelin levels were measured at baseline and weekly for 16 weeks. Ghrelin-expressing cells (GECs) in the stomach were assessed by using immunohistochemical staining and analyzed by using the Wilcoxon rank-sum test. Results In pigs treated with smaller microspheres, mean weight gain at 16 weeks (106.9% ± 15.0) was less than in control pigs (131.9% ± 11.6) (P < .001). Mean GEC density was lower in the gastric fundus (14.8 ± 6.3 vs 25.0 ± 6.9, P < .001) and body (27.5 ± 12.3 vs 37.9 ± 11.8, P = .004) but was not significantly different in the gastric antrum (28.2 ± 16.3 vs 24.3 ± 11.6, P = .84) and duodenum (9.2 ± 3.8 vs 8.7 ± 2.9, P = .66) versus in control pigs. BAE with larger microspheres failed to suppress weight gain or GECs in any stomach part compared with results in control swine. Plasma ghrelin levels were similar between BAE pigs and control pigs, regardless of microsphere size. Week 1 endoscopic evaluation for gastric ulcers revealed none in control pigs, five ulcers in five pigs embolized by using smaller microspheres, and three ulcers in five pigs embolized by using larger microspheres. Conclusion In bariatric arterial embolization, smaller microspheres rather than larger microspheres showed greater weight gain suppression and fundal ghrelin expression with more gastric ulceration in a swine model. © RSNA, 2018.


Assuntos
Embolização Terapêutica , Grelina/sangue , Aumento de Peso/fisiologia , Animais , Feminino , Artéria Gástrica/fisiologia , Grelina/metabolismo , Microesferas , Tamanho da Partícula , Estômago/irrigação sanguínea , Estômago/fisiologia , Suínos
6.
Theranostics ; 3(11): 916-26, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24396502

RESUMO

Poor cell survival and difficulties with visualization of cell delivery are major problems with current cell transplantation methods. To protect cells from early destruction, microencapsulation methods have been developed. The addition of a contrast agent to the microcapsule also could enable tracking by MR, ultrasound, and X-ray imaging. However, determining the cell viability within the microcapsule still remains an issue. Reporter gene imaging provides a way to determine cell viability, but delivery of the reporter probe by systemic injection may be hindered in ischemic diseases. In the present study, mesenchymal stem cells (MSCs) were transfected with triple fusion reporter gene containing red fluorescent protein, truncated thymidine kinase (SPECT/PET reporter) and firefly luciferase (bioluminescence reporter). Transfected cells were microencapsulated in either unlabeled or perfluorooctylbromide (PFOB) impregnated alginate. The addition of PFOB provided radiopacity to enable visualization of the microcapsules by X-ray imaging. Before intramuscular transplantation in rabbit thigh muscle, the microcapsules were incubated with D-luciferin, and bioluminescence imaging (BLI) was performed immediately. Twenty-four and forty-eight hours post transplantation, c-arm CT was used to target the luciferin to the X-ray-visible microcapsules for BLI cell viability assessment, rather than systemic reporter probe injections. Not only was the bioluminescent signal emission from the PFOB-encapsulated MSCs confirmed as compared to non-encapsulated, naked MSCs, but over 90% of injection sites of PFOB-encapsulated MSCs were visible on c-arm CT. The latter aided in successful targeting of the reporter probe to injection sites using conventional X-ray imaging to determine cell viability at 1-2 days post transplantation. Blind luciferin injections to the approximate location of unlabeled microcapsules resulted in successful BLI signal detection in only 18% of injections. In conclusion, reporter gene probes can be more precisely targeted using c-arm CT for in vivo transplant viability assessment, thereby avoiding large and costly systemic injections of a reporter probe.


Assuntos
Medições Luminescentes , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Imagem Molecular/métodos , Tomografia Computadorizada por Raios X/métodos , Animais , Genes Reporter , Luciferases de Vaga-Lume/análise , Luciferases de Vaga-Lume/genética , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Coelhos , Timidina Quinase/análise , Timidina Quinase/genética , Proteína Vermelha Fluorescente
7.
NMR Biomed ; 26(7): 850-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23225358

RESUMO

Microencapsulation of therapeutic cells has been widely pursued to achieve cellular immunoprotection following transplantation. Initial clinical studies have shown the potential of microencapsulation using semi-permeable alginate layers, but much needs to be learned about the optimal delivery route, in vivo pattern of engraftment, and microcapsule stability over time. In parallel with noninvasive imaging techniques for 'naked' (i.e. unencapsulated) cell tracking, microcapsules have now been endowed with contrast agents that can be visualized by (1) H MRI, (19) F MRI, X-ray/computed tomography and ultrasound imaging. By placing the contrast agent formulation in the extracellular space of the hydrogel, large amounts of contrast agents can be incorporated with negligible toxicity. This has led to a new generation of imaging biomaterials that can render cells visible with multiple imaging modalities.


Assuntos
Rastreamento de Células/métodos , Composição de Medicamentos/métodos , Animais , Cápsulas/química , Meios de Contraste/química , Dextranos/química , Humanos , Fenômenos Magnéticos , Nanopartículas de Magnetita/química
8.
Stem Cells ; 30(6): 1286-96, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22438076

RESUMO

The therapeutic goal in peripheral arterial disease (PAD) patients is to restore blood flow to ischemic tissue. Stem cell transplantation offers a new avenue to enhance arteriogenesis and angiogenesis. Two major problems with cell therapies are poor cell survival and the lack of visualization of cell delivery and distribution. To address these therapeutic barriers, allogeneic bone marrow-derived mesenchymal stem cells (MSCs) were encapsulated in alginate impregnated with a radiopaque contrast agent (MSC-Xcaps). In vitro MSC-Xcap viability by a fluorometric assay was high (96.9% ± 2.7% at 30 days postencapsulation) and as few as 10 Xcaps were visible on clinical x-ray fluoroscopic systems. Using an endovascular PAD model, rabbits (n = 21) were randomized to receive MSC-Xcaps (n = 6), empty Xcaps (n = 5), unencapsulated MSCs (n = 5), or sham intramuscular injections (n = 5) in the ischemic thigh 24 hours postocclusion. Immediately after MSC transplantation and 14 days later, digital radiographs acquired on a clinical angiographic system demonstrated persistent visualization of the Xcap injection sites with retained contrast-to-noise. Using a modified TIMI frame count, quantitative angiography demonstrated a 65% improvement in hind limb perfusion or arteriogenesis in MSC-Xcap-treated animals versus empty Xcaps. Post-mortem immunohistopathology of vessel density by anti-CD31 staining demonstrated an 87% enhancement in angiogenesis in Xcap-MSC-treated animals versus empty Xcaps. MSC-Xcaps represent the first x-ray-visible cellular therapeutic with enhanced efficacy for PAD treatment.


Assuntos
Membro Posterior/irrigação sanguínea , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Doença Arterial Periférica/cirurgia , Animais , Tomografia Computadorizada de Feixe Cônico/métodos , Modelos Animais de Doenças , Membro Posterior/patologia , Imuno-Histoquímica , Masculino , Células-Tronco Mesenquimais/citologia , Doença Arterial Periférica/fisiopatologia , Coelhos , Raios X
9.
Methods Mol Biol ; 680: 141-52, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21153379

RESUMO

Recently, several protocols for labeling of stem cells with superparamagnetic iron oxides (SPIOs) have been developed, leading to an active and growing field aimed at visualizing stem cells using MRI (magnetic resonance imaging), including image-guided stem cell injections. This development occurred simultaneously with a significant rise in the number of cell therapy clinical trials for cardiovascular applications and their preceding pre-clinical studies in animal models. In this chapter, we will describe several labeling strategies that can be used to label cells with SPIO nanoparticles. This is followed by a discussion of current strategies for using MRI to visualize these cells in myocardial infarct.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Eletroporação/métodos , Imageamento por Ressonância Magnética/métodos , Infarto do Miocárdio/patologia , Células-Tronco/ultraestrutura , Animais , Células Cultivadas , Meios de Contraste , Cães , Compostos Férricos , Infarto do Miocárdio/terapia , Polilisina , Protaminas , Transplante de Células-Tronco
10.
Methods Mol Biol ; 660: 171-83, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20680819

RESUMO

In the mid-1980s, iron oxide nanoparticles were developed as contrast agents for diagnostic imaging. In the last two decades, established methods to label cells with superparamagnetic iron oxides (SPIOs) have been developed to aid in targeted delivery and tracking of stem cell therapies. The surge in cellular therapy clinical trials for cardiovascular applications has seen a similar rise in the number of preclinical animal studies of SPIO-labeled stem cells in an effort to understand the mechanisms of cardiovascular regenerative therapy and stem cell biodistribution. The adoption of a limited number of methods of direct labeling of stem cells with SPIOs is due in large part to the desire to rapidly translate these techniques to clinical trials. In this review, we will outline the most commonly adopted methods for iron oxide labeling of stem cells for cardiovascular applications and describe strategies for magnetic resonance imaging (MRI) of magnetically labeled cells in the heart.


Assuntos
Doenças Cardiovasculares/terapia , Meios de Contraste/química , Óxido Ferroso-Férrico/química , Imageamento por Ressonância Magnética/métodos , Células-Tronco/citologia , Animais , Doenças Cardiovasculares/patologia , Cães , Eletroporação , Humanos , Suínos
11.
J Cardiovasc Transl Res ; 3(1): 24-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20376306

RESUMO

As clinical trials with stem cells for cardiac regenerative therapy move forward, advances in imaging equipment and technique offer powerful methods to evaluate therapeutic efficacy. Methodologies to label stem cells for tracking continue to expand. Non-invasive imaging offers the potential to better understand the interaction of exogenous stem cells with the host to answer questions such as the best cell type(s), timing of delivery, dose, and delivery route. If successful, these techniques may enable individually tailored dosing of stem cell therapeutics. However, techniques that are suitable for animal models of cardiac disease may have hurdles to clinical translation beyond simple biocompatibility issues. Challenges include the high cost of advanced imaging techniques, applicability in acute ischemic disease, and regulatory approval. In this review, we will cover some new imaging techniques and labeling strategies and assess the obstacles to clinical adoption.


Assuntos
Cardiopatias/cirurgia , Imagem Molecular , Miocárdio/patologia , Transplante de Células-Tronco , Animais , Meios de Contraste , Genes Reporter , Cardiopatias/diagnóstico , Cardiopatias/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Imagem Molecular/métodos , Imagem Molecular/tendências , Cintilografia , Regeneração , Tomografia Computadorizada por Raios X , Resultado do Tratamento
12.
Magn Reson Med ; 63(4): 1031-43, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20373404

RESUMO

With MRI (stem) cell tracking having entered the clinic, studies on the cellular genomic response toward labeling are warranted. Gene expression profiling was applied to C17.2 neural stem cells following superparamagnetic iron oxide/PLL (poly-L-lysine) labeling over the course of 1 week. Relative to unlabeled cells, less than 1% of genes (49 total) exhibited greater than 2-fold difference in expression in response to superparamagnetic iron oxide/PLL labeling. In particular, transferrin receptor 1 (Tfrc) and heme oxygenase 1 (Hmox1) expression was downregulated early, whereas genes involved in lysosomal function (Sulf1) and detoxification (Clu, Cp, Gstm2, Mgst1) were upregulated at later time points. Relative to cells treated with PLL only, cells labeled with superparamagnetic iron oxide/PLL complexes exhibited differential expression of 1399 genes. Though these differentially expressed genes exhibited altered expression over time, the overall extent was limited. Gene ontology analysis of differentially expressed genes showed that genes encoding zinc-binding proteins are enriched after superparamagnetic iron oxide/PLL labeling relative to PLL only treatment, whereas members of the apoptosis/programmed cell death pathway did not display increased expression. Overexpression of the differentially expressed genes Rnf138 and Abcc4 were confirmed by quantitative real-time polymerase chain reaction. These results demonstrate that, although early reactions responsible for iron homeostasis are induced, overall neural stem cell gene expression remains largely unaltered following superparamagnetic iron oxide/PLL labeling.


Assuntos
Meios de Contraste/química , Óxido Ferroso-Férrico/química , Perfilação da Expressão Gênica , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Algoritmos , Animais , Dextranos , Lisina/química , Nanopartículas de Magnetita , Células-Tronco Mesenquimais/metabolismo , Camundongos , RNA/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Magn Reson Med ; 60(6): 1506-11, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19025893

RESUMO

To optimize (19)F MR tracking of stem cells, we compared cellular internalization of cationic and anionic perfluoro-15-crown-5-ether (PFCE) nanoparticles using cell culture plates with different surface coatings. The viability and proliferation of anionic and cationic PFCE-labeled neural stem cells (NSCs) did not differ from unlabeled cells. Cationic PFCE nanoparticles ((19)F T1/T2 = 580/536 ms at 9.4 Tesla) were superior to anionic particles for intracellular fluorination. Best results were obtained with modified polystyrene culture dishes coated with both carboxylic and amino groups rather than conventional carboxyl-coated dishes. After injecting PFCE-labeled NSCs into the striatum of mouse brain, cells were readily identified in vivo by (19)F MRI without changes in signal or viability over a 2-week period after grafting. These results demonstrate that neural stem cells can be efficiently fluorinated with cationic PFCE nanoparticles without using transfection agents and visualized in vivo over prolonged periods with an MR sensitivity of approximately 140 pmol of PFCE/cell.


Assuntos
Radioisótopos de Flúor , Aumento da Imagem/métodos , Imageamento por Ressonância Magnética/métodos , Nanopartículas , Neurônios/citologia , Células-Tronco/citologia , Linhagem Celular , Portadores de Fármacos/química , Radioisótopos de Flúor/farmacocinética , Humanos , Nanopartículas/química , Neurônios/metabolismo , Coloração e Rotulagem/métodos , Células-Tronco/metabolismo
14.
J Am Coll Cardiol ; 52(6): 483-91, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18672170

RESUMO

OBJECTIVES: This study was designed to identify macrophage-rich atherosclerotic plaque noninvasively by imaging the tissue uptake of long-circulating superparamagnetic nanoparticles with a positive contrast off-resonance imaging sequence (inversion recovery with ON-resonant water suppression [IRON]). BACKGROUND: The sudden rupture of macrophage-rich atherosclerotic plaques can trigger the formation of an occlusive thrombus in coronary vessels, resulting in acute myocardial infarction. Therefore, a noninvasive technique that can identify macrophage-rich plaques and thereby assist with risk stratification of patients with atherosclerosis would be of great potential clinical utility. METHODS: Experiments were conducted on a clinical 3-T magnetic resonance imaging (MRI) scanner in 7 heritable hyperlipidemic and 4 control rabbits. Monocrystalline iron-oxide nanoparticles (MION)-47 were administrated intravenously (2 doses of 250 mumol Fe/kg), and animals underwent serial IRON-MRI before injection of the nanoparticles and serially after 1, 3, and 6 days. RESULTS: After administration of MION-47, a striking signal enhancement was found in areas of plaque only in hyperlipidemic rabbits. The magnitude of enhancement on magnetic resonance images had a high correlation with the number of macrophages determined by histology (p < 0.001) and allowed for the detection of macrophage-rich plaque with high accuracy (area under the curve: 0.92, SE: 0.04, 95% confidence interval: 0.84 to 0.96, p < 0.001). No significant signal enhancement was measured in remote areas without plaque by histology and in control rabbits without atherosclerosis. CONCLUSIONS: Using IRON-MRI in conjunction with superparamagnetic nanoparticles is a promising approach for the noninvasive evaluation of macrophage-rich, vulnerable plaques.


Assuntos
Arteriosclerose/diagnóstico , Meios de Contraste , Hiperlipidemias/complicações , Macrófagos , Imageamento por Ressonância Magnética/métodos , Tromboembolia/etiologia , Animais , Arteriosclerose/complicações , Arteriosclerose/patologia , Óxido Ferroso-Férrico , Macrófagos/patologia , Masculino , Nanopartículas , Coelhos , Medição de Risco
15.
J Magn Reson Imaging ; 27(5): 1175-80, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425827

RESUMO

PURPOSE: To investigate the utility of inversion recovery with ON-resonant water suppression (IRON) to create positive signal in normal lymph nodes after injection of superparamagnetic nanoparticles. MATERIALS AND METHODS: Experiments were conducted on six rabbits, which received a single bolus injection of 80 mumol Fe/kg monocrystalline iron oxide nanoparticle (MION-47). Magnetic resonance imaging (MRI) was performed at baseline, 1 day, and 3 days after MION-47 injection using conventional T(1)- and T(2)*-weighted sequences and IRON. Contrast-to-noise ratios (CNR) were measured in blood and in paraaortic lymph nodes. RESULTS: On T(2)*-weighted images, as expected, signal attenuation was observed in areas of paraaortic lymph nodes after MION-47 injection. However, using IRON the paraaortic lymph nodes exhibited very high contrast enhancement, which remained 3 days after injection. CNR with IRON was 2.2 +/- 0.8 at baseline, increased markedly 1 day after injection (23.5 +/- 5.4, P < 0.01 vs. baseline), and remained high after 3 days (21.8 +/- 5.7, *P < 0.01 vs. baseline). CNR was also high in blood 1 day after injection (42.7 +/- 7.2 vs. 1.8 +/- 0.7 at baseline, P < 0.01) but approached baseline after 3 days (1.9 +/- 1.4, P = NS vs. baseline). CONCLUSION: IRON in conjunction with superparamagnetic nanoparticles can be used to perform 'positive contrast' MR-lymphography, particularly 3 days after injection of the contrast agent, when signal is no longer visible within blood vessels. The proposed method may have potential as an adjunct for nodal staging in cancer screening.


Assuntos
Meios de Contraste/farmacocinética , Ferro/farmacocinética , Linfonodos/metabolismo , Imageamento por Ressonância Magnética/métodos , Óxidos/farmacocinética , Abdome , Análise de Variância , Animais , Meios de Contraste/administração & dosagem , Dextranos , Óxido Ferroso-Férrico , Processamento de Imagem Assistida por Computador , Injeções , Ferro/administração & dosagem , Nanopartículas de Magnetita , Masculino , Nanopartículas , Óxidos/administração & dosagem , Coelhos
16.
Stroke ; 39(5): 1569-74, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18323495

RESUMO

BACKGROUND AND PURPOSE: In animal models of stroke, functional improvement has been obtained after stem cell transplantation. Successful therapy depends largely on achieving a robust and targeted cell engraftment, with intraarterial (IA) injection being a potentially attractive route of administration. We assessed the suitability of laser Doppler flow (LDF) signal measurements and magnetic resonance (MR) imaging for noninvasive dual monitoring of targeted IA cell delivery. METHODS: Transient cerebral ischemia was induced in adult Wistar rats (n=25) followed by IA or intravenous (IV) injection of mesenchymal stem cells (MSCs) labeled with superparamagnetic iron oxide. Cell infusion was monitored in real time with transcranial laser Doppler flowmetry while cellular delivery was assessed with MRI in vivo (4.7 T) and ex vivo (9.4 T). RESULTS: Successful delivery of magnetically labeled MSCs could be readily visualized with MRI after IA but not IV injection. IA stem cell injection during acute stroke resulted in a high variability of cerebral engraftment. The amount of LDF reduction during cell infusion (up to 80%) was found to correlate well with the degree of intracerebral engraftment, with low LDF values being associated with significant morbidity. CONCLUSIONS: High cerebral engraftment rates are associated with impeded cerebral blood flow. Noninvasive dual-modality imaging enables monitoring of targeted cell delivery, and through interactive adjustment may improve the safety and efficacy of stem cell therapy.


Assuntos
Isquemia Encefálica/terapia , Artérias Cerebrais/cirurgia , Ataque Isquêmico Transitório/terapia , Fluxometria por Laser-Doppler/métodos , Imageamento por Ressonância Magnética/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Artérias Cerebrais/patologia , Artérias Cerebrais/fisiopatologia , Circulação Cerebrovascular/fisiologia , Feminino , Sobrevivência de Enxerto/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Monitorização Intraoperatória/métodos , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Acidente Vascular Cerebral/terapia , Resultado do Tratamento
17.
Magn Reson Med ; 58(5): 1072-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17969120

RESUMO

In proton magnetic resonance imaging (MRI) metallic substances lead to magnetic field distortions that often result in signal voids in the adjacent anatomic structures. Thus, metallic objects and superparamagnetic iron oxide (SPIO)-labeled cells appear as hypointense artifacts that obscure the underlying anatomy. The ability to illuminate these structures with positive contrast would enhance noninvasive MR tracking of cellular therapeutics. Therefore, an MRI methodology that selectively highlights areas of metallic objects has been developed. Inversion-recovery with ON-resonant water suppression (IRON) employs inversion of the magnetization in conjunction with a spectrally-selective on-resonant saturation prepulse. If imaging is performed after these prepulses, positive signal is obtained from off-resonant protons in close proximity to the metallic objects. The first successful use of IRON to produce positive contrast in areas of metallic spheres and SPIO-labeled stem cells in vitro and in vivo is presented.


Assuntos
Meios de Contraste , Compostos Férricos/química , Células-Tronco/química , Água/química
18.
Nanomedicine ; 2(2): 89-94, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17292120

RESUMO

Cellular magnetic resonance imaging (MRI) relies on the use of intracellular contrast agents, primarily iron oxide compounds. Several techniques have been used to efficiently shuttle iron oxides into nonphagocytic cells, but all methods used until now require a prolonged incubation of cells. We hypothesized that instant magnetic labeling of cells could be achieved using electroporation. Neural stem cells (NSCs) and leukocytes from spleen and lymph nodes were suspended in a ferumoxide labeling solution, loaded into cuvettes, and subjected to electromechanical permeabilization using electroporation. Magnetically labeled cells were assayed for labeling efficiency, as well as for potential toxicity or altered function. To confirm the method's applicability to detect cells, MRI experiments were performed at 11.7 T. Magnetoelectroporation of NSCs, as demonstrated by Prussian blue staining, anti-dextran immunostaining, and a quantitative iron uptake assay, proved to be an efficient intracellular magnetic labeling method. Leukocytes including lymphocytes, which are notoriously difficult to label because of their membrane properties and small cytoplasmic volume, also demonstrated a pronounced uptake of ferumoxide. MRI experiments showed that labeled NSCs could be visualized as single cells and cell clusters in gelatin phantoms, and as proliferating cell masses in mouse brain. We have developed a convenient technique for instant magnetic labeling of cells. Because magnetoelectroporation allows the use of ferumoxides approved by the US Food and Drug Administration without additional agents, it has excellent potential for clinical translation.


Assuntos
Eletroporação/métodos , Leucócitos/citologia , Imageamento por Ressonância Magnética/métodos , Magnetismo , Neurônios/citologia , Células-Tronco/citologia , United States Food and Drug Administration/legislação & jurisprudência , Animais , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Camundongos , Estados Unidos
19.
Circulation ; 112(10): 1451-61, 2005 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16129797

RESUMO

BACKGROUND: Recent results from animal studies suggest that stem cells may be able to home to sites of myocardial injury to assist in tissue regeneration. However, the histological interpretation of postmortem tissue, on which many of these studies are based, has recently been widely debated. METHODS AND RESULTS: With the use of the high sensitivity of a combined single-photon emission CT (SPECT)/CT scanner, the in vivo trafficking of allogeneic mesenchymal stem cells (MSCs) colabeled with a radiotracer and MR contrast agent to acute myocardial infarction was dynamically determined. Redistribution of the labeled MSCs after intravenous injection from initial localization in the lungs to nontarget organs such as the liver, kidney, and spleen was observed within 24 to 48 hours after injection. Focal and diffuse uptake of MSCs in the infarcted myocardium was already visible in SPECT/CT images in the first 24 hours after injection and persisted until 7 days after injection and was validated by tissue counts of radioactivity. In contrast, MRI was unable to demonstrate targeted cardiac localization of MSCs in part because of the lower sensitivity of MRI. CONCLUSIONS: Noninvasive radionuclide imaging is well suited to dynamically track the biodistribution and trafficking of mesenchymal stem cells to both target and nontarget organs.


Assuntos
Transplante de Células-Tronco Mesenquimais , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/terapia , Células-Tronco/diagnóstico por imagem , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Diferenciação Celular , Divisão Celular , Sobrevivência Celular , Cães , Radioisótopos de Índio , Injeções Intravenosas , Imageamento por Ressonância Magnética , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Compostos Organometálicos , Oxiquinolina/análogos & derivados , Reprodutibilidade dos Testes , Tomografia Computadorizada de Emissão de Fóton Único/normas , Tomografia Computadorizada por Raios X , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA