Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Arthritis Rheum ; 62(8): 2322-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20506388

ABSTRACT

OBJECTIVE: To evaluate a combined indocyanine green-enhanced optical imaging/radiography system for the detection of arthritic joints in a rat model of antigen-induced arthritis. METHODS: Arthritis of the knee and ankle joints was induced in 6 Harlan rats, using peptidoglycan-polysaccharide polymers. Three rats served as untreated controls. Optical imaging of the knee and ankle joints was done with an integrated optical imaging/radiography system before and up to 24 hours following intravenous injection of 10 mg/kg indocyanine green. The fluorescence signal intensities of arthritic and normal joints were compared for significant differences, using generalized estimating equation models. Specimens of knee and ankle joints were further processed and evaluated by histology. RESULTS: Immediately after administration, indocyanine green provided a significant increase in the fluorescence signal of arthritic joints compared with baseline values (P < 0.05). The fluorescence signal of arthritic joints was significantly higher compared with that of nonarthritic control joints at 1-720 minutes after intravenous injection (P < 0.05). Fusion of indocyanine green-enhanced optical imaging and radiography allowed for anatomic coregistration of the inflamed tissue with the associated joint. Hematoxylin and eosin staining confirmed marked synovial inflammation of arthritic joints and the absence of inflammation in control joints. CONCLUSION: Indocyanine green-enhanced optical imaging is a clinically applicable tool for detection of arthritic tissue. Using relatively high doses of indocyanine green, long-term enhanced fluorescence of arthritic joints can be achieved. This may facilitate simultaneous evaluations of multiple joints in a clinical setting. Fusion of indocyanine green-enhanced optical imaging scans with radiography increases anatomic resolution.


Subject(s)
Ankle Joint/diagnostic imaging , Arthritis, Experimental/diagnostic imaging , Arthrography/methods , Indocyanine Green , Knee Joint/diagnostic imaging , Animals , Radiographic Image Enhancement , Rats
2.
Pediatr Radiol ; 41(11): 1384-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21594541

ABSTRACT

BACKGROUND: Human embryonic stem cells (hESC) can generate cardiomyocytes (CM), which offer promising treatments for cardiomyopathies in children. However, challenges for clinical translation result from loss of transplanted cell from target sites and high cell death. An imaging technique that noninvasively and repetitively monitors transplanted hESC-CM could guide improvements in transplantation techniques and advance therapies. OBJECTIVE: To develop a clinically applicable labeling technique for hESC-CM with FDA-approved superparamagnetic iron oxide nanoparticles (SPIO) by examining labeling before and after CM differentiation. MATERIALS AND METHODS: Triplicates of hESC were labeled by simple incubation with 50 µg/ml of ferumoxides before or after differentiation into CM, then imaged on a 7T MR scanner using a T2-weighted multi-echo spin-echo sequence. Viability, iron uptake and T2-relaxation times were compared between groups using t-tests. RESULTS: hESC-CM labeled before differentiation demonstrated significant MR effects, iron uptake and preserved function. hESC-CM labeled after differentiation showed no significant iron uptake or change in MR signal (P < 0.05). Morphology, differentiation and viability were consistent between experimental groups. CONCLUSION: hESC-CM should be labeled prior to CM differentiation to achieve a significant MR signal. This technique permits monitoring delivery and engraftment of hESC-CM for potential advancements of stem cell-based therapies in the reconstitution of damaged myocardium.


Subject(s)
Contrast Media/metabolism , Embryonic Stem Cells/cytology , Magnetic Resonance Imaging , Myocytes, Cardiac/cytology , Cell Differentiation , Cell Survival , Cells, Cultured , Ferrosoferric Oxide/metabolism , Humans , Nanoparticles
3.
Mol Imaging ; 9(5): 278-90, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20868628

ABSTRACT

The purpose of this study was to compare viable and nonviable bilabeled mesenchymal stem cells (MSCs) in arthritic joints with magnetic resonance imaging (MRI) and optical imaging (OI). MSCs were labeled with ferucarbotran and DiD. MRI and OI of bilabeled cells were compared with controls. Six rats with arthritis received intra-articular injections of bilabeled viable MSCs into the right knee and nonviable MSCs into the left knee. Animals underwent MRI and OI preinjection and at 4, 24, 48, and 72 hours postinjection. The results were analyzed with a mixed random effects model and Fisher probability. Bilabeled MSCs showed increased MRI and OI signals compared to unlabeled controls (p < .0001). After intra-articular injection, bilabeled MSCs caused significant T2 and T2* effect on MRI and fluorescence on OI up to 72 hours postinjection (p < .05). There was no significant difference between viable and nonviable MSC signal in the knee joints; however, some of the viable cells migrated to an adjacent inflamed ankle joint (p < .05). Immunohistochemistry confirmed viable MSCs in right knee and ankle joints and nonviable MSCs in the left knee. Viable and nonviable cells could not be differentiated with MRI or OI signal intensity but were differentiated based on their ability to migrate in vivo.


Subject(s)
Magnetic Resonance Imaging/methods , Mesenchymal Stem Cells/cytology , Animals , Apoptosis/drug effects , Arthritis/therapy , Female , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mitomycin/pharmacology , Rats
4.
Radiology ; 255(2): 527-35, 2010 May.
Article in English | MEDLINE | ID: mdl-20413763

ABSTRACT

PURPOSE: To assess the capability of the folate receptor (FR)-targeted ultrasmall superparamagnetic iron oxide (USPIO) P1133 to provide FR-specific enhancement of breast cancers on magnetic resonance (MR) images. MATERIALS AND METHODS: This study was approved by the institutional Animal Care and Use Committee. The FR-targeted contrast agent P1133 was incubated with various FR-positive human breast cancer cell lines, with and without free folic acid (FFA) as a competitor. Labeling efficiencies were evaluated with MR imaging and inductively coupled plasma mass spectrometry. Subsequently, six athymic rats with implanted FR-positive MDA-MB-231 breast cancers underwent MR imaging at 3 T before and up to 1 hour and 24 hours after injection of P1133. Six athymic rats with implanted FR-positive MDA-MB-231 cancers injected with the non-FR-targeted USPIO P904 and nine athymic rats with implanted FR-negative A549 lung cancers injected with P1133 (n = 6) or P904 (n = 3) served as controls. Data of the in vitro studies were compared for significant differences with the Wilcoxon test for two independent samples. Tumor signal-to-noise-ratios (SNRs) were compared between different experimental groups by using the Kruskal-Wallis test and were correlated with histopathologic findings. Differences with P < .05 were considered significant. RESULTS: FR-positive breast cancer cells showed a significant P1133 uptake which was inhibited by FFA. MDA-MB-231 cells showed the highest level of P1133 uptake and the strongest T2 effect on MR images. In vivo, all tumors showed an initial perfusion effect. At 24 hours after injection, only MDA-MB-231 tumors injected with P1133 showed significantly decreased SNR data compared with baseline data (P < .05). MR findings were confirmed by using histopathologic findings. CONCLUSION: The FR-targeted USPIO P1133 demonstrates a specific retention in FR-positive breast cancers. Because FR expression correlates with tumor aggressiveness and prognosis, persistent P1133 tumor enhancement may be used as a noninvasive indicator for tumors with poor outcome.


Subject(s)
Breast Neoplasms/pathology , Contrast Media/chemistry , Dextrans/chemistry , Ferrosoferric Oxide/chemistry , Folic Acid/chemistry , Folic Acid/metabolism , Magnetic Resonance Imaging/methods , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , In Vitro Techniques , Magnetite Nanoparticles , Nanoparticles , Rats , Rats, Nude , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Statistics, Nonparametric , Transplantation, Heterologous
5.
J Transl Med ; 7: 94, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19906309

ABSTRACT

PURPOSE: Peri-tumoral inflammation is a common tumor response that plays a central role in tumor invasion and metastasis, and inflammatory cell recruitment is essential to this process. The purpose of this study was to determine whether injected fluorescently-labeled monocytes accumulate within murine breast tumors and are visible with optical imaging. MATERIALS AND METHODS: Murine monocytes were labeled with the fluorescent dye DiD and subsequently injected intravenously into 6 transgenic MMTV-PymT tumor-bearing mice and 6 FVB/n control mice without tumors. Optical imaging (OI) was performed before and after cell injection. Ratios of post-injection to pre-injection fluorescent signal intensity of the tumors (MMTV-PymT mice) and mammary tissue (FVB/n controls) were calculated and statistically compared. RESULTS: MMTV-PymT breast tumors had an average post/pre signal intensity ratio of 1.8+/- 0.2 (range 1.1-2.7). Control mammary tissue had an average post/pre signal intensity ratio of 1.1 +/- 0.1 (range, 0.4 to 1.4). The p-value for the difference between the ratios was less than 0.05. Confocal fluorescence microscopy confirmed the presence of DiD-labeled cells within the breast tumors. CONCLUSION: Murine monocytes accumulate at the site of breast cancer development in this transgenic model, providing evidence that peri-tumoral inflammatory cell recruitment can be evaluated non-invasively using optical imaging.


Subject(s)
Breast Neoplasms/pathology , Inflammation/pathology , Mammary Neoplasms, Experimental/pathology , Microscopy, Fluorescence/methods , Animals , Breast Neoplasms/immunology , Female , Fluorescent Dyes/metabolism , Humans , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Microscopy, Confocal/methods , Monocytes/cytology , Monocytes/metabolism
6.
Opt Express ; 17(26): 24403-13, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20052149

ABSTRACT

The objective of this work is to establish an optical imaging technique that would enable monitoring of the integration of mesenchymal stem cells (MSC) in arthritic joints. Our approach is based on first developing a labeling technique of MSC with the fluorescent dye DiD followed by tracking the cell migration kinetics from the spatial distribution of the DiD fluorescence in optical images (OI). The experimental approach involves first the in vitro OI of MSC labeled with DiD accompanied by fluorescence microscopy measurements to establish localization of the signal within the cells. Thereafter, DiD-labeled MSC were injected into polyarthritic, athymic rats and the signal localization within the experimental animals was monitored over several days. The experimental results indicate that DiD integrated into the cell membrane. DiD-labeled MSC localization in the arthritic ankle joints was observed with OI indicating that this method can be applied to monitor MSC in arthritic joints.


Subject(s)
Arthritis/immunology , Arthritis/pathology , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Animals , Cell Movement/immunology , Cells, Cultured , Disease Models, Animal , Female , Humans , Microscopy, Fluorescence , Rats , Rats, Nude
7.
Pediatr Radiol ; 39(11): 1194-202, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19763559

ABSTRACT

BACKGROUND: Long-term CT follow-up studies are required in pediatric patients who have received intraoperative radiation therapy (IORT) and external beam radiation therapy (EBRT) to assess vascular toxicities and to determine the exact complication rate. OBJECTIVE: To analyze with CT the effects of radiation therapy (RT) on the growth of the aorta in neuroblastoma patients. MATERIALS AND METHODS: Abdominal CT scans of 31 patients with intraabdominal neuroblastoma (stage II-IV), treated with RT (20 IORT+/-EBRT, 11 EBRT alone), were analyzed retrospectively. The diameter of the abdominal aorta was measured before and after RT. These data were compared to normal and predicted normal aortic diameters of children, according to the model of Fitzgerald, Donaldson and Poznanski (aortic diameter in centimeters = 0.844 + 0.0599 x age in years), and to the diameters of a control group of children who had not undergone RT. Statistical analyses for the primary aims were performed using the chi-squared test, t-test, Mann-Whitney test, nonparametric Wilcoxon matched-pairs test and analysis of variance for repeated measures. Clinical files and imaging studies were evaluated for signs of late vascular complications of neuroblastoma patients who had received RT. RESULTS: The mean diameter before and after RT and the growth of the aorta were significantly lower than expected in patients with neuroblastoma (P<0.05 for each) and when compared to the growth in a control group with normal and nonirradiated aortas. Among the patients who had received RT, there was no difference due to the type of RT. Seven patients from the IORT+/-EBRT group developed vascular complications, which included hypertension (five), middle aortic syndrome (two), death due to mesenteric ischemia (one) and critical aortic stenosis, which required aortic bypass surgery (two). CONCLUSION: Patients with neuroblastoma who had received RT showed impaired growth of the abdominal aorta. Significant long-term vascular complications occurred in seven patients who received IORT+/-EBRT. Thus, CT evaluation of patients with neuroblastoma who receive RT should include not only reports of changes in tumor extension, but also documentation of perfusion, and the size and growth of the aorta and its branches over time.


Subject(s)
Aorta/injuries , Aorta/radiation effects , Aortic Diseases/diagnostic imaging , Aortic Diseases/etiology , Neuroblastoma/radiotherapy , Radiation Injuries/diagnostic imaging , Radiation Injuries/etiology , Radiotherapy, Conformal/adverse effects , Aortography , Child , Child, Preschool , Female , Humans , Infant , Male , Neuroblastoma/complications , Retrospective Studies , Syndrome
8.
J Transl Med ; 6: 73, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-19038047

ABSTRACT

BACKGROUND: The development of postoperative granulation tissue is one of the main postoperative risks after lumbar spine surgery. This granulation tissue may lead to persistent or new clinical symptoms or complicate a follow up surgery. A sensitive non-invasive imaging technique, that could diagnose this granulation tissue at the bedside, would help to develop appropriate treatments. Thus, the purpose of this study was to establish a fast and economic imaging tool for the diagnosis of granulation tissue after lumbar spine surgery, using a new integrated Optical Imaging (OI)/X-ray imaging system and the FDA-approved fluorescent contrast agent Indocyanine Green (ICG). METHODS: 12 male Sprague Dawley rats underwent intervertebral disk surgery. Imaging of the operated lumbar spine was done with the integrated OI/X-ray system at 7 and 14 days after surgery. 6 rats served as non-operated controls. OI/X-ray scans of all rats were acquired before and after intravenous injection of the FDA-approved fluorescent dye Indocyanine Green (ICG) at a dose of 1 mg/kg or 10 mg/kg. The fluorescence signal of the paravertebral soft tissues was compared between different groups of rats using Wilcoxon-tests. Lumbar spines and paravertebral soft tissues were further processed with histopathology. RESULTS: In both dose groups, ICG provided a significant enhancement of soft tissue in the area of surgery, which corresponded with granulation tissue on histopathology. The peak and time interval of fluorescence enhancement was significantly higher using 10 mg/kg dose of ICG compared to the 1 mg/kg ICG dose. The levels of significance were p < 0.05. Fusion of OI data with X-rays allowed an accurate anatomical localization of the enhancing granulation tissue. CONCLUSION: ICG-enhanced OI is a suitable technique to diagnose granulation tissue after lumbar spine surgery. This new imaging technique may be clinically applicable for postoperative treatment monitoring. It could be also used to evaluate the effect of anti-inflammatory drugs and may even allow evaluations at the bedside with new hand-held OI scanners.


Subject(s)
Coloring Agents , Diagnostic Imaging/methods , Granulation Tissue , Indocyanine Green , Postoperative Complications , Animals , Granulation Tissue/diagnostic imaging , Granulation Tissue/pathology , Humans , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/pathology , Lumbar Vertebrae/surgery , Male , Postoperative Complications/diagnosis , Postoperative Complications/pathology , Postoperative Period , Radiography , Random Allocation , Rats , Rats, Sprague-Dawley , X-Rays
9.
AJR Am J Roentgenol ; 189(2): W96-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17646448

ABSTRACT

OBJECTIVE: The objective of our study was to determine the frequency and cause of anterior layering of excreted 18F-FDG in the bladder on PET/CT. CONCLUSION: Anterior layering of excreted FDG in the bladder is commonly seen on PET/CT scans obtained with i.v. iodinated contrast material and is due to displacement of FDG by excreted iodinated contrast material; this phenomenon may unmask FDG-avid bladder disease.


Subject(s)
Fluorodeoxyglucose F18/pharmacokinetics , Neoplasms/pathology , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Urinary Bladder Neoplasms/diagnostic imaging , Urinary Bladder Neoplasms/secondary , Urinary Bladder/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Contrast Media/administration & dosage , Female , Fluorodeoxyglucose F18/administration & dosage , Humans , Injections, Intravenous , Iohexol/administration & dosage , Male , Middle Aged , Neoplasms/diagnostic imaging , Phantoms, Imaging , Radiography , Radiopharmaceuticals/administration & dosage , Retrospective Studies , Tomography, Emission-Computed
10.
Acad Radiol ; 11(9): 1055-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15350587

ABSTRACT

RATIONALE AND OBJECTIVES: To assess the ability of magnetic resonance angiography (MRA) to evaluate complex vascular bypass reconstructions of the abdominal aorta and its major branches in the postoperative period. MATERIALS AND METHODS: Thirteen patients with bypass grafts connecting the aorta to visceral, renal, and lower limb inflow vessels were evaluated with MRA. Three of these patients were also studied with digital subtraction angiography soon after MRA was completed. MRA was evaluated for its ability to detect the grafts and to determine the degree of stenosis in the graft conduit or at the anastomoses to native vessels. RESULTS: Detection of graft conduits and anastomotic sites by MRA was 100% and 99%, respectively. Comparison with digital subtraction angiography in a subset of the patients showed a 100% agreement between the two modalities in their description of stenotic disease in graft conduits and 95% agreement in stenosis characterization at graft anastomotic sites. CONCLUSION: MRA of complex aortic reconstructions with bypass grafts to its major abdominal branches arteries accurately describes the resulting complicated vascular anatomy and likely has a high degree of correlation to digital subtraction angiography in describing the disease within the bypass grafts.


Subject(s)
Magnetic Resonance Angiography , Peripheral Vascular Diseases/surgery , Vascular Surgical Procedures , Viscera/blood supply , Viscera/diagnostic imaging , Anastomosis, Surgical , Angiography, Digital Subtraction , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/surgery , Celiac Artery/diagnostic imaging , Celiac Artery/surgery , Femoral Artery/diagnostic imaging , Femoral Artery/surgery , Graft Occlusion, Vascular/diagnosis , Graft Occlusion, Vascular/etiology , Humans , Mesenteric Artery, Superior/diagnostic imaging , Mesenteric Artery, Superior/surgery , Postoperative Complications/diagnosis , Postoperative Complications/etiology , Renal Artery/diagnostic imaging , Renal Artery/surgery , San Francisco , Severity of Illness Index , Treatment Outcome , Vascular Surgical Procedures/adverse effects , Viscera/surgery
11.
Cell Transplant ; 21(12): 2555-67, 2012.
Article in English | MEDLINE | ID: mdl-22862886

ABSTRACT

Magnetic resonance (MR) imaging of superparamagnetic iron oxide (SPIO)-labeled stem cells offers a noninvasive evaluation of stem cell engraftment in host organs. Excessive cellular iron load from SPIO labeling, however, impairs stem cell differentiation. The purpose of this study was to magnetically label human embryonic stem cells (hESCs) via a reduced exposure protocol that maintains a significant MR signal and no significant impairment to cellular pluripotency or differentiation potential. hESCs were labeled by simple incubation with Food and Drug Administration-approved ferumoxides, using concentrations of 50- 200 µg Fe/ml and incubation times of 3-24 h. The most reduced exposure labeling protocol that still provided a significant MR signal comparable to accepted labeling protocols was selected for subsequent studies. Labeled hESCs were compared to unlabeled controls for differences in pluripotency as studied by fluorescence staining for SSEA-1, SSEA-4, TRA-60, and TRA-81 and in differentiation capacity as studied by quantitative real-time PCR for hOCT4, hACTC1, hSOX1, and hAFP after differentiation into embryoid bodies (EBs). Subsequent MR and microscopy imaging were performed to evaluate for cellular iron distribution and long-term persistence of the label. An incubation concentration of 50 µg Fe/ml and incubation time of 3 h demonstrated a significantly reduced exposure protocol that yielded an intracellular iron uptake of 4.50 ± 0.27 pg, an iron content comparable to currently accepted SPIO labeling protocols. Labeled and unlabeled hESCs showed no difference in pluripotency or differentiation capacity. Ferumoxide-labeled hESCs demonstrated persistent MR contrast effects as embryoid bodies for 21 days. Electron microscopy confirmed persistent lysosomal storage of iron oxide particles in EBs up to 9 days, while additional microscopy visualization confirmed the iron distribution within single and multiple EBs. Labeling hESCs with ferumoxides by this tailored protocol reduces exposure of cells to the labeling agent while allowing for long-term visualization with MR imaging and the retention of cellular pluripotency and differentiation potential.


Subject(s)
Embryonic Stem Cells/cytology , Magnetics , Cell Differentiation , Cell Line , Contrast Media/chemistry , Contrast Media/metabolism , Dextrans/chemistry , Embryonic Stem Cells/metabolism , Humans , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Microscopy, Electron, Transmission
12.
Front Hum Neurosci ; 5: 111, 2011.
Article in English | MEDLINE | ID: mdl-22059071

ABSTRACT

Studies employing event-related potentials have shown that when participants are monitoring for a novel target face, the presentation of their own face elicits an enhanced negative brain potential in posterior channels approximately 250 ms after stimulus onset. Here, we investigate whether the own face N250 effect generalizes to other highly familiar objects, specifically, images of the participant's own dog and own car. In our experiments, participants were asked to monitor for a pre-experimentally unfamiliar target face (Joe), a target dog (Experiment 1: Joe's Dog) or a target car (Experiment 2: Joe's Car). The target face and object stimuli were presented with non-target foils that included novel face and object stimuli, the participant's own face, their own dog (Experiment 1), and their own car (Experiment 2). The consistent findings across the two experiments were the following: (1) the N250 potential differentiated the target faces and objects from the non-target face and object foils and (2) despite being non-targets, the own face and own objects produced an N250 response that was equal in magnitude to the target faces and objects by the end of the experiment. Thus, as indicated by its response to personally familiar and recently familiarized faces and objects, the N250 component is a sensitive index of individuated representations in visual memory.

13.
Mol Imaging Biol ; 13(1): 3-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20379785

ABSTRACT

PURPOSE: This study aims to determine the effect of human mesenchymal stem cell (hMSC) labeling with the fluorescent dye DiD and the iron oxide nanoparticle ferucarbotran on chondrogenesis. PROCEDURES: hMSCs were labeled with DiD alone or with DiD and ferucarbotran (DiD/ferucarbotran). hMSCs underwent confocal microscopy, optical imaging (OI), and magnetic resonance (MR) imaging. Chondrogenesis was induced by transforming growth factor-b and confirmed by histopathology and glycosaminoglycan (GAG) production. Data of labeled and unlabeled hMSCs were compared with a t test. RESULTS: Cellular uptake of DiD and ferucarbotran was confirmed with confocal microscopy. DiD labeling caused a significant fluorescence on OI, and ferucarbotran labeling caused a significant T2* effect on MR images. Compared to nonlabeled controls, progenies of labeled MSCs exhibited similar chondrocyte morphology after chondrogenic differentiation, but the labeled cells demonstrated significantly reduced GAG production (p < 0.05). CONCLUSION: DiD and DiD/ferucarbotran labeling of hMSC does not interfere with cell viability or morphologic differentiation into chondrocytes, but labeled cells exhibit significantly less GAG production compared to unlabeled cells.


Subject(s)
Contrast Media , Fluorescent Dyes , Mesenchymal Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Dextrans , Glycosaminoglycans/metabolism , Humans , Magnetic Resonance Imaging , Magnetite Nanoparticles , Mesenchymal Stem Cells/metabolism , Metal Nanoparticles
14.
Cell Transplant ; 19(1): 55-65, 2010.
Article in English | MEDLINE | ID: mdl-20370988

ABSTRACT

Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) have demonstrated the ability to improve myocardial function following transplantation into an ischemic heart; however, the functional benefits are transient possibly due to poor cell retention. A diagnostic technique that could visualize transplanted hESC-CMs could help to optimize stem cell delivery techniques. Thus, the purpose of this study was to develop a labeling technique for hESCs and hESC-CMs with the FDA-approved contrast agent indocyanine green (ICG) for optical imaging (OI). hESCs were labeled with 0.5, 1.0, 2.0, and 2.5 mg/ml of ICG for 30, 45, and 60 min at 37 degrees C. Longitudinal OI studies were performed with both hESCs and hESC-CMs. The expression of surface proteins was assessed with immunofluorescent staining. hESCs labeled with 2 mg ICG/ml for 60 min achieved maximum fluorescence. Longitudinal studies revealed that the fluorescent signal was equivalent to controls at 120 h postlabeling. The fluorescence signal of hESCs and hESC-CMs at 1, 24, and 48 h was significantly higher compared to precontrast data (p < 0.05). Immunocytochemistry revealed retention of cell-specific surface and nuclear markers postlabeling. These data demonstrate that hESCs and hESC-CMs labeled with ICG show a significant fluorescence up to 48 h and can be visualized with OI. The labeling procedure does not impair the viability or functional integrity of the cells. The technique may be useful for assessing different delivery routes in order to improve the engraftment of transplanted hESC-CMs or other stem cell progenitors.


Subject(s)
Embryonic Stem Cells/cytology , Fluorescent Antibody Technique/methods , Indocyanine Green/pharmacology , Myocytes, Cardiac/cytology , Staining and Labeling/methods , Stem Cell Transplantation/methods , Biomarkers/analysis , Biomarkers/metabolism , Cell Culture Techniques/methods , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Fluorescence , Heart Diseases/surgery , Humans , Membrane Proteins/analysis , Membrane Proteins/metabolism , Microscopy, Fluorescence/methods , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Time Factors
15.
Mol Imaging ; 8(1): 15-26, 2009.
Article in English | MEDLINE | ID: mdl-19344572

ABSTRACT

The purpose of this study was to track fluorophore-labeled, tumor-targeted natural killer (NK) cells to human prostate cancer xenografts with optical imaging (OI). NK-92-scFv(MOC31)-zeta cells targeted to the epithelial cell adhesion molecule (EpCAM) antigen on prostate cancer cells and nontargeted NK-92 parental cells were labeled with the near-infrared dye DiD (1,1'-dioctadecyl-3,3,3',3'-tetramethylindodicarbocyanine). The fluorescence, viability, and cytotoxicity of the labeled cells were evaluated. Subsequently, 12 athymic rats with prostate cancer xenografts underwent OI scans before and up to 24 hours postinjection of DiD-labeled parental NK-92 cells or NK-92-scFv(MOC31)-zeta cells. The tumor fluorescence intensity was measured and compared between pre- and postinjection scans and between both groups using t-tests. OI data were confirmed with fluorescence microscopy. In vitro studies demonstrated a significant increase in the fluorescence of labeled cells compared with unlabeled controls, which persisted over a period of 24 hours without any significant change in the viability. In vivo studies demonstrated a significant increase in tumor fluorescence at 24 hours postinjection of tumor-targeted NK-92-scFv(MOC31)-zeta cells but not parental NK cells. Ex vivo OI scans and fluorescence microscopy confirmed a specific accumulation of NK-92-scFv(MOC31)-zeta cells but not parental NK cells in the tumors. Tumor-targeted NK-92-scFv(MOC31)-zeta cells could be tracked to prostate cancer xenografts with OI.


Subject(s)
Immunotherapy, Adoptive , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Tomography, Optical/methods , Animals , CD3 Complex/immunology , Cytotoxicity, Immunologic/physiology , Fluorescent Dyes/pharmacology , Humans , Immunoglobulin Variable Region/metabolism , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Male , Microscopy, Fluorescence/methods , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Rats , Rats, Nude , Tumor Cells, Cultured
16.
J Vis Exp ; (13)2008 Mar 31.
Article in English | MEDLINE | ID: mdl-19066574

ABSTRACT

In recent years, stem cell research has led to a better understanding of developmental biology, various diseases and its potential impact on regenerative medicine. A non-invasive method to monitor the transplanted stem cells repeatedly in vivo would greatly enhance our ability to understand the mechanisms that control stem cell death and identify trophic factors and signaling pathways that improve stem cell engraftment. MR imaging has been proven to be an effective tool for the in vivo depiction of stem cells with near microscopic anatomical resolution. In order to detect stem cells with MR, the cells have to be labeled with cell specific MR contrast agents. For this purpose, iron oxide nanoparticles, such as superparamagnetic iron oxide particles (SPIO), are applied, because of their high sensitivity for cell detection and their excellent biocompatibility. SPIO particles are composed of an iron oxide core and a dextran, carboxydextran or starch coat, and function by creating local field inhomogeneities, that cause a decreased signal on T2-weighted MR images. This presentation will demonstrate techniques for labeling of stem cells with clinically applicable MR contrast agents for subsequent non-invasive in vivo tracking of the labeled cells with MR imaging.


Subject(s)
Embryonic Stem Cells/cytology , Ferric Compounds/chemistry , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cells/cytology , Nanoparticles/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Embryonic Stem Cells/metabolism , Ferric Compounds/metabolism , Humans , Mesenchymal Stem Cells/metabolism
17.
Pediatr Radiol ; 38(5): 529-37, 2008 May.
Article in English | MEDLINE | ID: mdl-18357444

ABSTRACT

BACKGROUND: Because of its over-expression in many human tumors, the folate receptor (FR) is a promising target for tumor-specific imaging. OBJECTIVE: To evaluate the uptake of FR-targeted gadolinium (P866) and iron-oxide (P1048) agents in an ovarian tumor model. MATERIALS AND METHODS: FR-positive ovarian cancer cells (IGROV-1) were incubated with FR-targeted agents (P866 or P1048) in the absence or presence of competing free folate. Intracellular gadolinium or iron-oxide concentrations were measured. MR imaging of implanted ovarian tumors in rats was performed following injection of FR-targeted (P866 and P1048) and nontargeted (P1001 and P904) agents. Changes in longitudinal and transverse relaxation rates (DeltaR1 and DeltaR2), which were proportional to the contrast agent concentration in the tumors, were compared between tumors injected with FR-targeted and nontargeted agents. RESULTS: IGROV-1 cells showed uptake of P866 and P1048, which decreased with competing free folate. The DeltaR1 values were higher at 1 h following injection of P866 than following injection of P1001 (P < 0.05), indicating a higher amount of contrast agent retained in the tumor following P866 injection. There was a trend for higher DeltaR2 values at 48 h following injection of P1048 than following injection of P904, but it was not statistically significant (P = 0.09). CONCLUSION: Specific accumulation of the FR-targeted gadolinium agent P866 was suggested in an FR-positive ovarian tumor model, demonstrating the possibility of combining the specificity of receptor targeting with the improved anatomic resolution of MR imaging. This could improve diagnosis and treatment of FR-positive tumors.


Subject(s)
Carrier Proteins , Contrast Media/administration & dosage , Magnetic Resonance Imaging/methods , Ovarian Neoplasms/diagnosis , Ovary/pathology , Animals , Disease Models, Animal , Feasibility Studies , Female , Ferric Compounds , Folate Receptors, GPI-Anchored , Gadolinium , Humans , Image Enhancement/methods , Image Processing, Computer-Assisted/methods , Ovarian Neoplasms/pathology , Rats , Rats, Nude , Receptors, Cell Surface , Sensitivity and Specificity , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
J Vis Exp ; (14)2008 Apr 02.
Article in English | MEDLINE | ID: mdl-19066580

ABSTRACT

Optical imaging (OI) is an easy, fast and inexpensive tool for in vivo monitoring of new stem cell based therapies. The technique is based on ex vivo labeling of stem cells with a fluorescent dye, subsequent intravenous injection of the labeled cells and visualization of their accumulation in specific target organs or pathologies. The presented technique demonstrates how we label human mesenchymal stem cells (hMSC) by simple incubation with the lipophilic fluorescent dye DiD (C67H103CIN2O3S) and how we label human embryonic stem cells (hESC) with the FDA approved fluorescent dye Indocyanine Green (ICG). The uptake mechanism is via adherence and diffusion of the lypophilic dye across the phospholipid cell membrane bilayer. The labeling efficiency is usually improved if the cells are incubated with the dye in serum-free media as opposed to incubation in serum-containing media. Furthermore, the addition of the transfection agent Protamine Sulfate significantly improves contrast agent uptake.


Subject(s)
Fluorescent Dyes , Mesenchymal Stem Cells/cytology , Animals , Follow-Up Studies , Humans , Mesenchymal Stem Cell Transplantation/methods , Microscopy, Fluorescence , Rats , Reproducibility of Results , Staining and Labeling/methods
19.
Contrast Media Mol Imaging ; 3(5): 191-7, 2008.
Article in English | MEDLINE | ID: mdl-18973215

ABSTRACT

Indocyanine green (ICG) is a contrast agent used for detecting angiogenesis with optical imaging (OI). The purpose of this study was to investigate whether cooling procedures increase the signal yield of ICG with OI. Test samples of 0.05 and 0.02 mM ICG in 40% DMSO and 60% DMEM underwent OI at four different temperatures (5, 37, 55 and 75 degrees C). In addition, six athymic rats with an antigen-induced arthritis of the knee and ankle joints underwent OI before and after injection of ICG (10 mg/ml, dose 15 mg/kg) on two separate days with and without cooling of the joints. The fluorescent signals of the test samples and arthritic joints were measured and evaluated for significant differences before and after cooling with a t-test. In vitro studies showed a strong negative correlation between ICG temperature and fluorescent signal. The mean fluorescent signal of arthritic joints (measured in efficiency) was 0.345 before ICG-injection, 4.55 after ICG-injection and before cooling and 9.71 after ICG-injection and after cooling. The fluorescent signal enhancement of arthritic joints with ICG-enhanced OI images increased significantly after cooling (p = 0.02). The signal yield of ICG can be significantly increased by cooling the target pathology. The primary underlying cause of the temperature dependence of ICG is enhanced collisional quenching with increasing temperature. This simple cooling method may be immediately helpful to increase the fluorescence signal yield in current ICG-enhanced OI-studies in patients.


Subject(s)
Arthritis, Experimental/diagnosis , Cold Temperature , Fluorescence , Indocyanine Green/chemistry , Animals , Ankle Joint/pathology , Coloring Agents/chemistry , Contrast Media/chemistry , Diagnostic Imaging/methods , Female , Knee Joint/pathology , Rats , Rats, Nude
20.
Eur Radiol ; 17(5): 1226-34, 2007 May.
Article in English | MEDLINE | ID: mdl-17206428

ABSTRACT

The purpose of this study was to label human monocytes with Gadofluorine M by simple incubation for subsequent cell depiction at 1.5 and 3 T. Gadofluorine M displays a high r(1) relaxivity and is spontaneously phagocytosed by macrophages. Human monocytes were incubated with Gadofluorine M-Cy at varying concentrations and incubation times and underwent MR imaging at 1.5 and 3 T at increasing time intervals after the labeling procedure. R1-relaxation rates and r1 relaxivities of the labeled cells and non-labeled controls were determined. Cellular contrast agent uptake was examined by fluorescence microscopy and quantified by ICP-AES. Efficient cell labeling was achieved after incubation of the cells with 25 mM Gd Gadofluorine M for 12 h, resulting in a maximal uptake of 0.3 fmol Gd/cell without impairment of cell viability. Fluorescence microscopy confirmed internalization of the fluorescent contrast agent by monocytes. The r1 relaxivity of the labeled cells was 137 mM(-1)s(-1) at 1.5 T and 80.46 mM(-1)s(-1) at 3 T. Imaging studies showed stable labeling for at least 7 days. Human monocytes can be effectively labeled for MR imaging with Gadofluorine M. Potential in vivo cell-tracking applications include targeting of inflammatory processes with Gadofluorine-labeled leukocytes or monitoring of stem cell therapies for the treatment of arthritis.


Subject(s)
Contrast Media/pharmacokinetics , Magnetic Resonance Imaging/methods , Monocytes/metabolism , Organometallic Compounds/pharmacokinetics , Cells, Cultured , Fluorocarbons , Humans , Microscopy, Confocal , Spectrophotometry, Atomic , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL