Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Theranostics ; 8(17): 4837-4855, 2018.
Article in English | MEDLINE | ID: mdl-30279741

ABSTRACT

Magnetic resonance imaging (MRI)-guided pulsed focused ultrasound (pFUS) combined with microbubbles (MB) contrast agent infusion has been shown to transiently disrupt the blood-brain barrier (BBBD), increasing the delivery of neurotherapeutics to treat central nervous system (CNS) diseases. pFUS interaction with the intravascular MB results in acoustic cavitation forces passing through the neurovascular unit (NVU), inducing BBBD detected on contrast-enhanced MRI. Multiple pFUS+MB exposures in Alzheimer's disease (AD) models are being investigated as a method to clear amyloid plaques by activated microglia or infiltrating immune cells. Since it has been reported that pFUS+MB can induce a sterile inflammatory response (SIR) [1-5] in the rat, the goal of this study was to investigate the potential long-term effects of SIR in the brain following single and six weekly sonications by serial high-resolution MRI and pathology. Methods: Female Sprague Dawley rats weighing 217±16.6 g prior to sonication received bromo-deoxyuridine (BrdU) to tag proliferating cells in the brain. pFUS was performed at 548 kHz, ultrasound burst 10 ms and initial peak negative pressure of 0.3 MPa (in water) for 120 s coupled with a slow infusion of ~460 µL/kg (5-8×107 MB) that started 30 s before and 30 s during sonication. Nine 2 mm focal regions in the left cortex and four regions over the right hippocampus were treated with pFUS+MB. Serial high-resolution brain MRIs at 3 T and 9.4 T were obtained following a single or during the course of six weekly pFUS+MB resulting in BBBD in the left cortex and the right hippocampus. Animals were monitored over 7 to 13 weeks and imaging results were compared to histology. Results: Fewer than half of the rats receiving a single pFUS+MB exposure displayed hypointense voxels on T2*-weighted (w) MRI at week 7 or 13 in the cortex or hippocampus without differences compared to the contralateral side on histograms of T2* maps. Single sonicated rats had evidence of limited microglia activation on pathology compared to the contralateral hemisphere. Six weekly pFUS+MB treatments resulted in pathological changes on T2*w images with multiple hypointense regions, cortical atrophy, along with 50% of rats having persistent BBBD and astrogliosis by MRI. Pathologic analysis of the multiple sonicated animals demonstrated the presence of metallophagocytic Prussian blue-positive cells in the parenchyma with significantly (p<0.05) increased areas of activated astrocytes and microglia, and high numbers of systemic infiltrating CD68+ macrophages along with BrdU+ cells compared to contralateral brain. In addition, multiple treatments caused an increase in the number of hyperphosphorylated Tau (pTau)-positive neurons containing neurofibrillary tangles (NFT) in the sonicated cortex but not in the hippocampus when compared to contralateral brain, which was confirmed by Western blot (WB) (p<0.04). Conclusions: The repeated SIR following multiple pFUS+MB treatments could contribute to changes on MR imaging including persistent BBBD, cortical atrophy, and hypointense voxels on T2w and T2*w images consistent with pathological injury. Moreover, areas of astrogliosis, activated microglia, along with higher numbers of CD68+ infiltrating macrophages and BrdU+ cells were detected in multiple sonicated areas of the cortex and hippocampus. Elevations in pTau and NFT were detected in neurons of the multiple sonicated cortex. Minimal changes on MRI and histology were observed in single pFUS+MB-treated rats at 7 and 13 weeks post sonication. In comparison, animals that received 6 weekly sonications demonstrated evidence on MRI and histology of vascular damage, inflammation and neurodegeneration associated with the NVU commonly observed in trauma. Further investigation is recommended of the long-term effects of multiple pFUS+MB in clinical trials.


Subject(s)
Cerebral Cortex/pathology , Cerebral Cortex/radiation effects , Hippocampus/pathology , Hippocampus/radiation effects , Microbubbles/adverse effects , Ultrasonography/adverse effects , Animals , Histocytochemistry , Longitudinal Studies , Magnetic Resonance Imaging , Rats, Sprague-Dawley
2.
J Transl Med ; 15(1): 252, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29237455

ABSTRACT

BACKGROUND: Image-guided high intensity focused ultrasound has been used as an extracorporeal cardiac pacing tool and to enhance homing of stem cells to targeted tissues. However, molecular changes in the myocardium after sonication have not been widely investigated. Magnetic-resonance (MR)-guided pulsed focused ultrasound (pFUS) was targeted to the rat myocardium over a range of pressures and the microenvironmental and histological effects were evaluated over time. METHODS: Eight-to-ten-week-old Sprague-Dawley rats received T2-weighted MR images to target pFUS to the left ventricular and septum without cardiac or respiratory gating. Rats were sonicated through the thoracic wall at peak negative pressures (PNP) from 1 to 8 MPa at a center frequency of 1 MHz, 10 ms pulse duration and 1 Hz pulse repetition frequency for 100 pulses per focal target. Following pFUS, myocardium was harvested over 24 h and subjected to imaging, proteomic, and histological measurements. RESULTS: pFUS to the myocardium increased expression of cytokines, chemokines, and trophic factors characterized by an initial increase in tumor necrosis factor (TNF)-α followed by increases in pro- and anti-inflammatory factors that returned to baseline by 24 h. Immediately after pFUS, there was a transient (< 1 h) increase in N-terminal pro b-type natriuretic peptide (NT-proBNP) without elevation of other cardiac injury markers. A relationship between PNP and expression of TNF-α and NT-proBNP was observed with significant changes (p < 0.05 ANOVA) ≥ 4 MPa compared to untreated controls. Contrast-enhanced ex vivo T1-weighted MRI revealed vascular leakage in sonicated myocardium that was accompanied by the presence of albumin upon immunohistochemistry. Histology revealed infiltration of neutrophils and macrophages without morphological myofibril changes in sonicated tissue accompanied by pulmonary hemorrhage at PNP > 4 MPa. CONCLUSIONS: MR-guided pFUS to myocardium induced transient proteomic and histological changes. The temporal proteomic changes in the myocardium indicate a short-lived sterile inflammatory response consistent with ischemia or contusion. Further study of myocardial function and strain is needed to determine if pFUS could be developed as an experimental model of cardiac injury and chest trauma.


Subject(s)
Heart/diagnostic imaging , High-Intensity Focused Ultrasound Ablation , Magnetic Resonance Imaging , Myocardium/metabolism , Myocardium/pathology , Animals , Biomarkers/metabolism , Female , Granulocytes/metabolism , Macrophages/metabolism , Proteomics , Rats, Sprague-Dawley
3.
Sci Transl Med ; 9(374)2017 01 25.
Article in English | MEDLINE | ID: mdl-28123074

ABSTRACT

Long-term neurological deficits due to immature cortical development are emerging as a major challenge in congenital heart disease (CHD). However, cellular mechanisms underlying dysregulation of perinatal corticogenesis in CHD remain elusive. The subventricular zone (SVZ) represents the largest postnatal niche of neural stem/progenitor cells (NSPCs). We show that the piglet SVZ resembles its human counterpart and displays robust postnatal neurogenesis. We present evidence that SVZ NSPCs migrate to the frontal cortex and differentiate into interneurons in a region-specific manner. Hypoxic exposure of the gyrencephalic piglet brain recapitulates CHD-induced impaired cortical development. Hypoxia reduces proliferation and neurogenesis in the SVZ, which is accompanied by reduced cortical growth. We demonstrate a similar reduction in neuroblasts within the SVZ of human infants born with CHD. Our findings demonstrate that SVZ NSPCs contribute to perinatal corticogenesis and suggest that restoration of SVZ NSPCs' neurogenic potential is a candidate therapeutic target for improving cortical growth in CHD.


Subject(s)
Frontal Lobe/pathology , Heart Failure/pathology , Neurogenesis , Animals , Animals, Newborn , Cell Movement , Frontal Lobe/growth & development , Heart Failure/congenital , Hypoxia/pathology , Interneurons/cytology , Neural Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology , Stem Cell Niche , Stem Cells/cytology , Swine
4.
Proc Natl Acad Sci U S A ; 114(1): E75-E84, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27994152

ABSTRACT

MRI-guided pulsed focused ultrasound (pFUS) combined with systemic infusion of ultrasound contrast agent microbubbles (MB) causes localized blood-brain barrier (BBB) disruption that is currently being advocated for increasing drug or gene delivery in neurological diseases. The mechanical acoustic cavitation effects of opening the BBB by low-intensity pFUS+MB, as evidenced by contrast-enhanced MRI, resulted in an immediate damage-associated molecular pattern (DAMP) response including elevations in heat-shock protein 70, IL-1, IL-18, and TNFα indicative of a sterile inflammatory response (SIR) in the parenchyma. Concurrent with DAMP presentation, significant elevations in proinflammatory, antiinflammatory, and trophic factors along with neurotrophic and neurogenesis factors were detected; these elevations lasted 24 h. Transcriptomic analysis of sonicated brain supported the proteomic findings and indicated that the SIR was facilitated through the induction of the NFκB pathway. Histological evaluation demonstrated increased albumin in the parenchyma that cleared by 24 h along with TUNEL+ neurons, activated astrocytes, microglia, and increased cell adhesion molecules in the vasculature. Infusion of fluorescent beads 3 d before pFUS+MB revealed the infiltration of CD68+ macrophages at 6 d postsonication, as is consistent with an innate immune response. pFUS+MB is being considered as part of a noninvasive adjuvant treatment for malignancy or neurodegenerative diseases. These results demonstrate that pFUS+MB induces an SIR compatible with ischemia or mild traumatic brain injury. Further investigation will be required before this approach can be widely implemented in clinical trials.


Subject(s)
Blood-Brain Barrier/physiology , Brain/physiopathology , Drug Delivery Systems/methods , Gene Transfer Techniques , Sonication/methods , Ultrasonography/methods , Animals , Astrocytes/metabolism , Cell Adhesion Molecules/metabolism , Female , HSP70 Heat-Shock Proteins/metabolism , Inflammation/pathology , Interleukin-1/metabolism , Interleukin-18/metabolism , Macrophages/immunology , Microglia/metabolism , Neurodegenerative Diseases/therapy , Parenchymal Tissue/pathology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
5.
Nanomedicine ; 13(2): 503-513, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27520728

ABSTRACT

Stem cell-based therapies have become a major focus in regenerative medicine and to treat diseases. A straightforward approach combining three drugs, heparin (H), protamine (P) with ferumoxytol (F) in the form of nanocomplexes (NCs) effectively labeled stem cells for cellular MRI. We report on the physicochemical characteristics for optimizing the H, P, and F components in different ratios, and mixing sequences, producing NCs that varied in hydrodynamic size. NC size depended on the order in which drugs were mixed in media. Electron microscopy of HPF or FHP showed that F was located on the surface of spheroidal shaped HP complexes. Human stem cells incubated with FHP NCs resulted in a significantly greater iron concentration per cell compared to that found in HPF NCs with the same concentration of F. These results indicate that FHP could be useful for labeling stem cells in translational studies in the clinic.


Subject(s)
Ferrosoferric Oxide , Heparin , Protamines , Stem Cells , Cell Tracking , Humans , Magnetic Resonance Imaging , Magnetics , Nanoparticles , Stem Cell Transplantation
6.
PLoS One ; 10(5): e0126551, 2015.
Article in English | MEDLINE | ID: mdl-25946089

ABSTRACT

Mesenchymal stromal cells secrete a variety of anti-inflammatory factors and may provide a regenerative medicine option for the treatment of traumatic brain injury. The present study investigates the efficacy of multiple intravenous or intracardiac administrations of rat mesenchymal stromal cells or human mesenchymal stromal cells in female rats after controlled cortical impact by in vivo MRI, neurobehavior, and histopathology evaluation. Neither intravenous nor intracardiac administration of mesenchymal stromal cells derived from either rats or humans improved MRI measures of lesion volume or neurobehavioral outcome compared to saline treatment. Few mesenchymal stromal cells (<0.0005% of injected dose) were found within 3 days of last dosage at the site of injury after either delivery route, with no mesenchymal stromal cells being detectable in brain at 30 or 56 days post-injury. These findings suggest that non-autologous mesenchymal stromal cells therapy via intravenous or intracardiac administration is not a promising treatment after focal contusion traumatic brain injury in this female rodent model.


Subject(s)
Brain Injuries/therapy , Brain/pathology , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Administration, Intravenous , Animals , Brain/cytology , Cardiac Catheters , Cells, Cultured , Disease Models, Animal , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Mesenchymal Stem Cells/cytology , Rats , Rats, Wistar , Treatment Failure
7.
Bioorg Med Chem Lett ; 25(10): 2056-9, 2015.
Article in English | MEDLINE | ID: mdl-25870133

ABSTRACT

There is growing interest in small and rigid peptidomimetic αvß3 integrin antagonists that are readily synthesized and characterized and amenable to physiological conditions. Peptidomimetic 4-[2-(3,4,5,6-tetrahydropyrimidine-2-ylamino)ethyloxy]benzoyl-2-[N-(3-amino-neopenta-1-carbamyl)]-aminoethylsulfonyl-amino-ß-alanine (IAC) was successfully conjugated to DOTA, complexed with Gd(III) and radiolabeled with (153)Gd. Radioassay results demonstrated specificity of the labeled conjugate by blocking ∼95% binding with the addition of a 50-fold molar excess of cold IAC to the reaction solution. Relaxometry was used to support the hypothesis that the specificity of the Gd-peptidomimetic targeting αvß3 integrin would increase the contrast and therefore enhance the sensitivity of an MRI scan of αvß3 integrin positive tissues. Magnetic resonance imaging of cell pellets (M21 human melanoma) was also performed, and the images clearly show that cells reacted with Gd(III)-DOTA-IAC display a brighter image than cells without the Gd(III)-DOTA-IAC contrast agent. In addition, Gd(III)-DOTA-IAC and IAC, with IC50 of 300nM and 230nM, respectively, are 2.1 and 2.7 times more potent than c(RGDfK) whose IC50 is 625nM. This promising preliminary data fuels further investigation of DOTA-IAC conjugates for targeting tumor associated angiogenesis and αvß3 integrin positive tumors using magnetic resonance imaging.


Subject(s)
Contrast Media/chemical synthesis , Coordination Complexes/chemical synthesis , Gadolinium/chemistry , Integrin alphaVbeta3/chemistry , Magnetic Resonance Imaging , Peptidomimetics/chemistry , Cell Line, Tumor , Chromatography, High Pressure Liquid , Contrast Media/chemistry , Coordination Complexes/chemistry , Humans , Inhibitory Concentration 50 , Peptidomimetics/chemical synthesis
8.
NMR Biomed ; 26(4): 468-79, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23225324

ABSTRACT

Serial MRI facilitates the in vivo analysis of the intra- and intersubject evolution of traumatic brain injury lesions. Despite the availability of MRI, the natural history of experimental focal contusion lesions in the controlled cortical impact (CCI) rat model has not been well described. We performed CCI on rats and MRI during the acute to chronic stages of cerebral injury to investigate the time course of changes in the brain. Female Wistar rats underwent CCI of their left motor cortex with a flat impact tip driven by an electromagnetic piston. In vivo MRI was performed at 7 T serially over 6 weeks post-CCI. The appearances of CCI-induced lesions and lesion-associated cortical volumes were variable on MRI, with the percentage change in cortical volume of the CCI ipsilateral side relative to the contralateral side ranging from 18% within 2 h of injury on day 0 to a peak of 35% on day 1, and a trough of -28% by week 5/6, with an average standard deviation of ± 14% at any given time point. In contrast, the percentage change in cortical volume of the ipsilateral side relative to the contralateral side in control rats was not significant (1 ± 2%). Hemorrhagic conversion within and surrounding the CCI lesion occurred between days 2 and 9 in 45% of rats, with no hemorrhage noted on the initial scan. Furthermore, hemorrhage and hemosiderin within the lesion were positive for Prussian blue and highly autofluorescent on histological examination. Although some variation in injuries may be technique related, the divergence of similar lesions between initial and final scans demonstrates the inherent biological variability of the CCI rat model.


Subject(s)
Brain Injuries/complications , Brain Injuries/pathology , Contusions/complications , Contusions/pathology , Animals , Behavior, Animal , Cerebral Cortex/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Disease Models, Animal , Female , Magnetic Resonance Imaging , Microscopy, Fluorescence , Organ Size , Rats, Wistar
9.
Stem Cells ; 30(6): 1216-27, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22593018

ABSTRACT

Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/diagnostic imaging , Bone Marrow Transplantation/methods , Kidney/cytology , Kidney/diagnostic imaging , Stromal Cells/transplantation , Ultrasonics/methods , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/diagnostic imaging , Cell Culture Techniques , Cytokines/metabolism , Female , Humans , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/diagnostic imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Stromal Cells/cytology , Ultrasonography
10.
Nat Med ; 18(3): 463-7, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366951

ABSTRACT

We report on a new straightforward magnetic cell-labeling approach that combines three US Food and Drug Administration (FDA)-approved drugs--ferumoxytol, heparin and protamine--in serum-free medium to form self-assembling nanocomplexes that effectively label cells for in vivo magnetic resonance imaging (MRI). We observed that the ferumoxytol-heparin-protamine (HPF) nanocomplexes were stable in serum-free cell culture medium. HPF nanocomplexes show a threefold increase in T2 relaxivity compared to ferumoxytol. Electron microscopy showed internalized HPF in endosomes, which we confirmed by Prussian blue staining of labeled cells. There was no long-term effect or toxicity on cellular physiology or function of HPF-labeled hematopoietic stem cells, bone marrow stromal cells, neural stem cells or T cells when compared to controls. In vivo MRI detected 1,000 HPF-labeled cells implanted in rat brains. This HPF labeling method should facilitate the monitoring by MRI of infused or implanted cells in clinical trials.


Subject(s)
Brain/cytology , Cell Tracking/methods , Ferrosoferric Oxide/chemistry , Heparin/chemistry , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/adverse effects , Magnetite Nanoparticles/chemistry , Protamines/chemistry , Animals , Apoptosis/drug effects , Bone Marrow Cells/cytology , Brain/diagnostic imaging , Brain/ultrastructure , Cell Differentiation/drug effects , Clinical Trials as Topic , Culture Media, Serum-Free , Endosomes/ultrastructure , Ferrosoferric Oxide/administration & dosage , Ferrosoferric Oxide/adverse effects , Hematopoietic Stem Cells/cytology , Heparin/administration & dosage , Heparin/adverse effects , Humans , Magnetic Resonance Imaging , Magnetite Nanoparticles/ultrastructure , Male , Mesenchymal Stem Cells/cytology , Microscopy, Electron , Neurons/cytology , Protamines/administration & dosage , Protamines/adverse effects , Radiography , Rats , Stem Cell Transplantation , Stromal Cells/cytology , T-Lymphocytes/cytology
11.
PLoS One ; 6(9): e24730, 2011.
Article in English | MEDLINE | ID: mdl-21931834

ABSTRACT

Continuous focused ultrasound (cFUS) has been widely used for thermal ablation of tissues, relying on continuous exposures to generate temperatures necessary to induce coagulative necrosis. Pulsed FUS (pFUS) employs non-continuous exposures that lower the rate of energy deposition and allow cooling to occur between pulses, thereby minimizing thermal effects and emphasizing effects created by non-thermal mechanisms of FUS (i.e., acoustic radiation forces and acoustic cavitation). pFUS has shown promise for a variety of applications including drug and nanoparticle delivery; however, little is understood about the effects these exposures have on tissue, especially with regard to cellular pro-homing factors (growth factors, cytokines, and cell adhesion molecules). We examined changes in murine hamstring muscle following pFUS or cFUS and demonstrate that pFUS, unlike cFUS, has little effect on the histological integrity of muscle and does not induce cell death. Infiltration of macrophages was observed 3 and 8 days following pFUS or cFUS exposures. pFUS increased expression of several cytokines (e.g., IL-1α, IL-1ß, TNFα, INFγ, MIP-1α, MCP-1, and GMCSF) creating a local cytokine gradient on days 0 and 1 post-pFUS that returns to baseline levels by day 3 post-pFUS. pFUS exposures induced upregulation of other signaling molecules (e.g., VEGF, FGF, PlGF, HGF, and SDF-1α) and cell adhesion molecules (e.g., ICAM-1 and VCAM-1) on muscle vasculature. The observed molecular changes in muscle following pFUS may be utilized to target cellular therapies by increasing homing to areas of pathology.


Subject(s)
High-Intensity Focused Ultrasound Ablation , Animals , Apoptosis/physiology , Cell Adhesion Molecules/metabolism , Chemokine CCL3/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1/metabolism , Interleukin-1beta/metabolism , Macrophages , Magnetic Resonance Imaging , Mice , Muscle, Skeletal/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
12.
NMR Biomed ; 24(3): 325-34, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20949637

ABSTRACT

This study uses quantitative T(2)* imaging to track ferumoxides--protamine sulfate (FEPro)-labeled MDA-MB-231BR-Luc (231BRL) human breast cancer cells that metastasize to the nude rat brain. Four cohorts of nude rats were injected intracardially with FEPro-labeled, unlabeled or tumor necrosis factor-related apoptosis-inducing ligand(TRAIL)-treated (to induce apoptosis) 231BRL cells, or saline, in order to develop metastatic breast cancer in the brain. The heads of the rats were imaged serially over 3-4 weeks using gradient multi-echo and turbo spin-echo pulse sequences at 3 T with a solenoid receive-only 4-cm-diameter coil. Quantitative T(2)* maps of the whole brain were obtained by the application of single-exponential fitting to the signal intensity of T(2)* images, and the distribution of T(2)* values in brain voxels was calculated. MRI findings were correlated with Prussian blue staining and immunohistochemical staining for iron in breast cancer and macrophages. Quantitative analysis of T(2)* from brain voxels demonstrated a significant shift to lower values following the intracardiac injection of FEPro-labeled 231BRL cells, relative to animals receiving unlabeled cells, apoptotic cells or saline. Quartile analysis based on the T(2)* distribution obtained from brain voxels demonstrated significant differences (p < 0.0083) in the number of voxels with T(2)* values in the ranges 10-35 ms (Q1), 36-60 ms (Q2) and 61-86 ms (Q3) from 1 day to 3 weeks post-infusion of labeled 231BRL cells, compared with baseline scans. There were no significant differences in the distribution of T(2)* obtained from serial MRI in rats receiving unlabeled or TRAIL-treated cells or saline. Histologic analysis demonstrated isolated Prussian blue-positive breast cancer cells scattered in the brains of rats receiving labeled cells, relative to animals receiving unlabeled or apoptotic cells. Quantitative T(2)* analysis of FEPro-labeled metastasized cancer cells was possible even after the hypointense voxels were no longer visible on T(2)*-weighted images.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Magnetic Resonance Imaging/methods , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Dextrans/metabolism , Female , Humans , Magnetite Nanoparticles , Neoplasm Metastasis , Neoplasm Transplantation , Protamines/metabolism , Rats , Rats, Nude
13.
J Transl Med ; 7: 88, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19840404

ABSTRACT

BACKGROUND: Establishing a large rodent model of brain metastasis that can be monitored using clinically relevant magnetic resonance imaging (MRI) techniques is challenging. Non-invasive imaging of brain metastasis in mice usually requires high field strength MR units and long imaging acquisition times. Using the brain seeking MDA-MB-231BR transfected with luciferase gene, a metastatic breast cancer brain tumor model was investigated in the nude rat. Serial MRI and bioluminescence imaging (BLI) was performed and findings were correlated with histology. Results demonstrated the utility of multimodality imaging in identifying unexpected sights of metastasis and monitoring the progression of disease in the nude rat. METHODS: Brain seeking breast cancer cells MDA-MB-231BR transfected with firefly luciferase (231BRL) were labeled with ferumoxides-protamine sulfate (FEPro) and 1-3 x 106 cells were intracardiac (IC) injected. MRI and BLI were performed up to 4 weeks to monitor the early breast cancer cell infiltration into the brain and formation of metastases. Rats were euthanized at different time points and the imaging findings were correlated with histological analysis to validate the presence of metastases in tissues. RESULTS: Early metastasis of the FEPro labeled 231BRL were demonstrated on T2*-weighted MRI and BLI within 1 week post IC injection of cells. Micro-metastatic tumors were detected in the brain on T2-weighted MRI as early as 2 weeks post-injection in greater than 85% of rats. Unexpected skeletal metastases from the 231BRL cells were demonstrated and validated by multimodal imaging. Brain metastases were clearly visible on T2 weighted MRI by 3-4 weeks post infusion of 231BRL cells, however BLI did not demonstrate photon flux activity originating from the brain in all animals due to scattering of the photons from tumors. CONCLUSION: A model of metastatic breast cancer in the nude rat was successfully developed and evaluated using multimodal imaging including MRI and BLI providing the ability to study the temporal and spatial distribution of metastases in the brain and skeleton.


Subject(s)
Breast Neoplasms/pathology , Luminescent Measurements/methods , Magnetic Resonance Imaging/methods , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis , Animals , Bone Neoplasms/secondary , Brain/anatomy & histology , Brain/pathology , Brain Neoplasms/secondary , Cell Line, Tumor , Dextrans , Female , Ferrosoferric Oxide/chemistry , Ferrosoferric Oxide/metabolism , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Magnetite Nanoparticles , Mice , Protamines/chemistry , Protamines/metabolism , Rats
14.
PLoS One ; 4(8): e6712, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19696933

ABSTRACT

Intracellular labels such as dextran coated superparamagnetic iron oxide nanoparticles (SPION), bromodeoxyuridine (BrdU) or green fluorescent protein (GFP) are frequently used to study the fate of transplanted cells by in vivo magnetic resonance imaging or fluorescent microscopy. Bystander uptake of labeled cells by resident tissue macrophages (TM) can confound the interpretation of the presence of intracellular labels especially during direct implantation of cells, which can result in more than 70% cell death. In this study we determined the percentages of TM that took up SPION, BrdU or GFP from labeled bone marrow stromal cells (BMSCs) that were placed into areas of angiogenesis and inflammation in a mouse model known as Matrigel plaque perfusion assay. Cells recovered from digested plaques at various time points were analyzed by fluorescence microscopy and flow cytometry. The analysis of harvested plaques revealed 5% of BrdU(+), 5-10% of GFP(+) and 5-15% of dextran(+) macrophages. The transfer of the label was not dependent on cell dose or viability. Collectively, this study suggests that care should be taken to validate donor origin of cells using an independent marker by histology and to assess transplanted cells for TM markers prior to drawing conclusions about the in vivo behavior of transplanted cells.


Subject(s)
Bone Marrow Cells/cytology , Macrophages/cytology , Stromal Cells/cytology , Animals , Bromodeoxyuridine/metabolism , Cell Lineage , Flow Cytometry , Green Fluorescent Proteins/genetics , Humans , Magnetic Resonance Imaging , Mice , Microscopy, Fluorescence
15.
NMR Biomed ; 18(6): 383-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16013087

ABSTRACT

Ferumoxides, dextran-coated superparamagnetic iron oxide (SPIO) particles, form ferumoxide-transfection agent (FE-TA) complexes that are internalized into endosomes/lysosomes and have been used to label cells for in vivo MRI tracking and localization studies. A better understanding of the physical state of the FE-TA complexes during endocytosis could improve their use. The purpose of this study was to measure the rate of the degradation of iron particles under varying physiological conditions. FE-TA complexes were incubated in seven different buffers containing different chelates with different pH. Reducible iron concentrations, T2 relaxation rates and gradient echo (GRE) magnetic resonance images (MRI) were obtained from each condition immediately after incubation and at 6, 24, 48, 72 and 96 h and days 7, 14 and 21. The dynamics of FE-TA in the endosome/lysosomes within the cells were visualized with electron microscopy. Sodium citrate buffer at pH 4.5 rapidly dissolved FE-TA complexes. However, FE-TA complexes were less soluble in the same buffer at pH 5.5. Similarly, FE-TA complexes were not readily soluble in any of the other buffers with or without chelates, regardless of pH. Electron microscopic images showed degraded FE-TA in some intracellular endosome/lysosomes between days 3 and 5. In the cellular environment, some of the FE-TA-containing endosomes were found to fuse with lysosomes, causing rapid dissociation at low pH and exposing the iron core to chelates that resulted in soluble Fe(III) within the lysosomes. The studies presented represent a first step in identifying the important cellular environmental parameters affecting the integrity of FE-TA complexes.


Subject(s)
Iron/chemistry , Iron/pharmacokinetics , Lysosomes/metabolism , Lysosomes/ultrastructure , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cells/metabolism , Nanotubes/chemistry , Oxides/chemistry , Oxides/pharmacokinetics , Cell Separation/methods , Cells, Cultured , Coated Materials, Biocompatible/pharmacokinetics , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Dextrans/chemistry , Ferrosoferric Oxide , Humans , Magnetite Nanoparticles , Metabolic Clearance Rate , Nanotubes/ultrastructure , Staining and Labeling/methods
16.
Hum Gene Ther ; 15(4): 351-60, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15053860

ABSTRACT

Targeted delivery of intravenously administered genetically altered cells or stem cells is still in an early stage of investigation. We developed a method of delivering iron oxide (ferumoxide)-labeled mesenchymal stem cells (MSCs) to a targeted area in an animal model by applying an external magnet. Rats with or without an external magnet placed over the liver were injected intravenously with ferumoxide-labeled MSCs and magnetic resonance imaging (MRI) signal intensity (SI) changes, iron concentration, and concentration of MSCs in the liver were monitored at different time points. SI decreased in the liver after injection of MSCs and returned gradually to that of control rat livers at approximately day 29. SI decreases were greater in rats with external magnets. Higher iron concentration and increased labeled cell numbers were detected in rat livers with external magnets. The external magnets influenced the movement of labeled MSCs such that the cells were retained in the region of interest. These results potentially open a new area of investigation for delivering stem cells or genetically altered cells.


Subject(s)
Biological Transport , Drug Delivery Systems , Liver/pathology , Magnetic Resonance Imaging/methods , Magnetics , Mesenchymal Stem Cell Transplantation/methods , Animals , Cells, Cultured , Dextrans , Ferrosoferric Oxide , Iron , Magnetite Nanoparticles , Mesoderm/cytology , Oxides , Rats , Rats, Nude
17.
Radiology ; 229(3): 838-46, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14657318

ABSTRACT

PURPOSE: To evaluate the effect of using the ferumoxides-poly-l-lysine (PLL) complex for magnetic cell labeling on the long-term viability, function, metabolism, and iron utilization of mammalian cells. MATERIALS AND METHODS: PLL was incubated with ferumoxides for 60 minutes, incompletely coating the superparamagnetic iron oxide (SPIO) through electrostatic interactions. Cells were coincubated overnight with the ferumoxides-PLL complex, and iron uptake, cell viability, apoptosis indexes, and reactive oxygen species formation were evaluated. The disappearance or the life span of the detectable iron nanoparticles in cells was also evaluated. The iron concentrations in the media also were assessed at different time points. Data were expressed as the mean +/- 1 SD, and one-way analysis of variance and the unpaired Student t test were used to test for significant differences. RESULTS: Intracytoplasmic nanoparticles were stained with Prussian blue when the ferumoxides-PLL complex had magnetically labeled the human mesenchymal stem and HeLa cells. The long-term viability, growth rate, and apoptotic indexes of the labeled cells were unaffected by the endosomal incorporation of SPIO, as compared with these characteristics of the nonlabeled cells. In nondividing human mesenchymal stem cells, endosomal iron nanoparticles could be detected after 7 weeks; however, in rapidly dividing cells, intracellular iron had disappeared by five to eight divisions. A nonsignificant transient increase in reactive oxygen species production was seen in the human mesenchymal stem and HeLa cell lines. Labeled human mesenchymal stem cells did not differentiate to other lineage. A significant increase in iron concentration was observed in both the human mesenchymal stem and HeLa cell media at day 7. CONCLUSION: Magnetic cellular labeling with the ferumoxides-PLL complex had no short- or long-term toxic effects on tumor or stem cells.


Subject(s)
Contrast Media , HeLa Cells/physiology , Iron/pharmacokinetics , Magnetic Resonance Imaging , Oxides/pharmacokinetics , Stem Cells/physiology , Apoptosis , Cell Survival , Cells, Cultured , Contrast Media/pharmacokinetics , Contrast Media/toxicity , Dextrans , Ferrocyanides , Ferrosoferric Oxide , HeLa Cells/metabolism , Humans , Iron/toxicity , Magnetite Nanoparticles , Oxides/toxicity , Particle Size , Polylysine/pharmacokinetics , Polylysine/toxicity , Reactive Oxygen Species/metabolism , Stem Cells/metabolism
18.
Radiology ; 228(2): 480-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12819345

ABSTRACT

PURPOSE: To label mammalian and stem cells by combining commercially available transfection agents (TAs) with superparamagnetic iron oxide (SPIO) magnetic resonance (MR) imaging contrast agents. MATERIALS AND METHODS: Three TAs were incubated with ferumoxides and MION-46L in cell culture medium at various concentrations. Human mesenchymal stem cells, mouse lymphocytes, rat oligodendrocyte progenitor CG-4 cells, and human cervical carcinoma cells were incubated 2-48 hours with 25 microg of iron per milliliter of combined TAs and SPIO. Cellular labeling was evaluated with T2 relaxometry, MR imaging of labeled cell suspensions, and Prussian blue staining for iron assessment. Proliferation and viability of mesenchymal stem cells and human cervical carcinoma cells labeled with a combination of TAs and ferumoxides were evaluated. RESULTS: When ferumoxides-TA or MION-46L-TA was used, intracytoplasmic particles stained with Prussian blue stain were detected for all cell lines with a labeling efficiency of nearly 100%. Limited or no uptake was observed for cells incubated with ferumoxides or MION-46L alone. For TA-SPIO-labeled cells, MR images and relaxometry findings showed a 50%-90% decrease in signal intensity and a more than 40-fold increase in T2s. Cell viability varied from 103.7% +/- 9 to 123.0% +/- 9 compared with control cell viability at 9 days, and cell proliferation was not affected by endosomal incorporation of SPIO nanoparticles. Iron concentrations varied with ferumoxides-TA combinations and cells with a maximum of 30.1 pg +/- 3.7 of iron per cell for labeled mesenchymal stem cells. CONCLUSION: Magnetic labeling of mammalian cells with use of ferumoxides and TAs is possible and may enable cellular MR imaging and tracking in experimental and clinical settings.


Subject(s)
Indicators and Reagents/pharmacokinetics , Iron/pharmacokinetics , Lipids/pharmacokinetics , Magnetic Resonance Imaging , Oxides/pharmacokinetics , Polylysine/pharmacokinetics , Transfection/methods , Animals , Cells, Cultured , Female , Ferrosoferric Oxide , Humans , Liposomes , Mice , Rats , Stem Cells/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...