Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Molecules ; 28(16)2023 Aug 13.
Article in English | MEDLINE | ID: mdl-37630292

ABSTRACT

In the field of nuclear medicine, the ß+ -emitting 43Sc and ß- -emitting 47Sc are promising candidates in cancer diagnosis and targeted radionuclide therapy (TRT) due to their favorable decay schema and shared pharmacokinetics as a true theranostic pair. Additionally, scandium is a group-3 transition metal (like 177Lu) and exhibits affinity for DOTA-based chelators, which have been studied in depth, making the barrier to implementation lower for 43/47Sc than for other proposed true theranostics. Before 43/47Sc can see widespread pre-clinical evaluation, however, an accessible production methodology must be established and each isotope's radiolabeling and animal imaging capabilities studied with a widely utilized tracer. As such, a simple means of converting an 18 MeV biomedical cyclotron to support solid targets and produce 43Sc via the 42Ca(d,n)43Sc reaction has been devised, exhibiting reasonable yields. The NatTi(γ,p)47Sc reaction is also investigated along with the successful implementation of chemical separation and purification methods for 43/47Sc. The conjugation of 43/47Sc with PSMA-617 at specific activities of up to 8.94 MBq/nmol and the subsequent imaging of LNCaP-ENZaR tumor xenografts in mouse models with both 43/47Sc-PSMA-617 are also presented.


Subject(s)
Nuclear Medicine , Prostatic Neoplasms , Humans , Animals , Mice , Male , Scandium , Precision Medicine , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Radioisotopes/therapeutic use
2.
Nanomaterials (Basel) ; 13(4)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36839041

ABSTRACT

Photodynamic therapy (PDT), the use of light to excite photosensitive molecules whose electronic relaxation drives the production of highly cytotoxic reactive oxygen species (ROS), has proven an effective means of oncotherapy. However, its application has been severely constrained to superficial tissues and those readily accessed either endoscopically or laparoscopically, due to the intrinsic scattering and absorption of photons by intervening tissues. Recent advances in the design of nanoparticle-based X-ray scintillators and photosensitizers have enabled hybridization of these moieties into single nanocomposite particles. These nanoplatforms, when irradiated with diagnostic doses and energies of X-rays, produce large quantities of ROS and permit, for the first time, non-invasive deep tissue PDT of tumors with few of the therapeutic limitations or side effects of conventional PDT. In this review we examine the underlying principles and evolution of PDT: from its initial and still dominant use of light-activated, small molecule photosensitizers that passively accumulate in tumors, to its latest development of X-ray-activated, scintillator-photosensitizer hybrid nanoplatforms that actively target cancer biomarkers. Challenges and potential remedies for the clinical translation of these hybrid nanoplatforms and X-ray PDT are also presented.

3.
Eur J Nucl Med Mol Imaging ; 49(12): 4014-4024, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35792927

ABSTRACT

PURPOSE: To identify the optimal threshold in 18F-fluoromisonidazole (FMISO) PET images to accurately locate tumor hypoxia by using electron paramagnetic resonance imaging (pO2 EPRI) as ground truth for hypoxia, defined by pO2 [Formula: see text] 10 mmHg. METHODS: Tumor hypoxia images in mouse models of SCCVII squamous cell carcinoma (n = 16) were acquired in a hybrid PET/EPRI imaging system 2 h post-injection of FMISO. T2-weighted MRI was used to delineate tumor and muscle tissue. Dynamic contrast enhanced (DCE) MRI parametric images of Ktrans and ve were generated to model tumor vascular properties. Images from PET/EPR/MRI were co-registered and resampled to isotropic 0.5 mm voxel resolution for analysis. PET images were converted to standardized uptake value (SUV) and tumor-to-muscle ratio (TMR) units. FMISO uptake thresholds were evaluated using receiver operating characteristic (ROC) curve analysis to find the optimal FMISO threshold and unit with maximum overall hypoxia similarity (OHS) with pO2 EPRI, where OHS = 1 shows perfect overlap and OHS = 0 shows no overlap. The means of dice similarity coefficient, normalized Hausdorff distance, and accuracy were used to define the OHS. Monotonic relationships between EPRI/PET/DCE-MRI were evaluated with the Spearman correlation coefficient ([Formula: see text]) to quantify association of vasculature on hypoxia imaged with both FMISO PET and pO2 EPRI. RESULTS: FMISO PET thresholds to define hypoxia with maximum OHS (both OHS = 0.728 [Formula: see text] 0.2) were SUV [Formula: see text] 1.4 [Formula: see text] SUVmean and SUV [Formula: see text] 0.6 [Formula: see text] SUVmax. Weak-to-moderate correlations (|[Formula: see text]|< 0.70) were observed between PET/EPRI hypoxia images with vascular permeability (Ktrans) or fractional extracellular-extravascular space (ve) from DCE-MRI. CONCLUSION: This is the first in vivo comparison of FMISO uptake with pO2 EPRI to identify the optimal FMISO threshold to define tumor hypoxia, which may successfully direct hypoxic tumor boosts in patients, thereby enhancing tumor control.


Subject(s)
Carcinoma, Squamous Cell , Tumor Hypoxia , Animals , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/pathology , Cell Hypoxia , Electron Spin Resonance Spectroscopy , Hypoxia/diagnostic imaging , Mice , Misonidazole/analogs & derivatives , Positron-Emission Tomography/methods , Radiopharmaceuticals , Tomography, X-Ray Computed
4.
Radiol Imaging Cancer ; 3(2): e200104, 2021 03.
Article in English | MEDLINE | ID: mdl-33817651

ABSTRACT

Purpose: To enhance the spatial accuracy of fluorine 18 (18F) misonidazole (MISO) PET imaging of hypoxia by using dynamic contrast-enhanced (DCE) MR images as a basis for modifying PET images and by using electron paramagnetic resonance (EPR) partial oxygen pressure (pO2) as the reference standard. Materials and Methods: Mice (n = 10) with leg-borne MCa4 mammary carcinomas underwent EPR imaging, T2-weighted and DCE MRI, and 18F-MISO PET/CT. Images were registered to the same space for analysis. The thresholds of hypoxia for PET and EPR images were tumor-to-muscle ratios greater than or equal to 2.2 mm Hg and less than or equal to 14 mm Hg, respectively. The Dice similarity coefficient (DSC) and Hausdorff distance (d H ) were used to quantify the three-dimensional overlap of hypoxia between pO2 EPR and 18F-MISO PET images. A training subset (n = 6) was used to calculate optimal DCE MRI weighting coefficients to relate EPR to the PET signal; the group average weights were then applied to all tumors (from six training mice and four test mice). The DSC and d H were calculated before and after DCE MRI-corrected PET images were obtained to quantify the improvement in overlap with EPR pO2 images for measuring tumor hypoxia. Results: The means and standard deviations of the DSC and d H between hypoxic regions in original PET and EPR images were 0.35 mm ± 0.23 and 5.70 mm ± 1.7, respectively, for images of all 10 mice. After implementing a preliminary DCE MRI correction to PET data, the DSC increased to 0.86 mm ± 0.18 and the d H decreased to 2.29 mm ± 0.70, showing significant improvement (P < .001) for images of all 10 mice. Specifically, for images of the four independent test mice, the DSC improved with correction from 0.19 ± 0.28 to 0.80 ± 0.29 (P = .02), and the d H improved from 6.40 mm ± 2.5 to 1.95 mm ± 0.63 (P = .01). Conclusion: Using EPR information as a reference standard, DCE MRI information can be used to correct 18F-MISO PET information to more accurately reflect areas of hypoxia.Keywords: Animal Studies, Molecular Imaging, Molecular Imaging-Cancer, PET/CT, MR-Dynamic Contrast Enhanced, MR-Imaging, PET/MR, Breast, Oncology, Tumor Mircoenvironment, Electron Paramagnetic ResonanceSupplemental material is available for this article.© RSNA, 2021.


Subject(s)
Misonidazole , Tumor Hypoxia , Animals , Electron Spin Resonance Spectroscopy , Hypoxia/diagnostic imaging , Magnetic Resonance Imaging , Mice , Oxygen , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography
5.
Neurocrit Care ; 34(1): 64-72, 2021 02.
Article in English | MEDLINE | ID: mdl-32358767

ABSTRACT

BACKGROUND: Cardiac arrest (CA) patients who survived by cardiopulmonary resuscitation (CPR) can present different levels of neurological deficits ranging from minor cognitive impairments to persistent vegetative state and brain death. The pathophysiology of the resulting brain injury is poorly understood, and whether changes in post-CA brain metabolism contribute to the injury are unknown. Here we utilized [18F]fluorodeoxyglucose (FDG)-Positron emission tomography (PET) to study in vivo cerebral glucose metabolism 72 h following CA in a murine CA model. METHODS: Anesthetized and ventilated adult C57BL/6 mice underwent 12-min KCl-induced CA followed by CPR. Seventy-two hours following CA, surviving mice were intraperitoneally injected with [18F]FDG (~ 186 µCi/200 µL) and imaged on Molecubes preclinical micro-PET/computed tomography (CT) imaging systems after a 30-min awake uptake period. Brain [18F]FDG uptake was determined by the VivoQuant software on fused PET/CT images with the 3D brain atlas. Upon completion of Positron emission tomography (PET) imaging, remaining [18F]FDG radioactivity in the brain, heart, and liver was determined using a gamma counter. RESULTS: Global increases in brain [18F]FDG uptake in post-CA mice were observed compared to shams and controls. The median standardized uptake value of [18F]FDG for CA animals was 1.79 versus sham 1.25 (p < 0.05) and control animals 0.78 (p < 0.01). This increased uptake was consistent throughout the 60-min imaging period and across all brain regions reaching statistical significance in the midbrain, pons, and medulla. Biodistribution analyses of various key organs yielded similar observations that the median [18F]FDG uptake for brain was 7.04%ID/g tissue for CA mice versus 5.537%ID/g tissue for sham animals, p < 0.05). CONCLUSIONS: This study has successfully applied [18F]FDG-PET/CT to measure changes in brain metabolism in a murine model of asystolic CA. Our results demonstrate increased [18F]FDG uptake in the brain 72 h following CA, suggesting increased metabolic demand in the case of severe neurological injury. Further study is warranted to determine the etiology of these changes.


Subject(s)
Fluorodeoxyglucose F18 , Heart Arrest , Animals , Brain/diagnostic imaging , Glucose , Heart Arrest/diagnostic imaging , Humans , Mice , Mice, Inbred C57BL , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography , Radiopharmaceuticals , Tissue Distribution
6.
Angew Chem Int Ed Engl ; 59(35): 15161-15165, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32415874

ABSTRACT

Herein, we report the development of an 18 F-labeled, activity-based small-molecule probe targeting the cancer-associated serine hydrolase NCEH1. We undertook a focused medicinal chemistry campaign to simultaneously preserve potent and specific NCEH1 labeling in live cells and animals, while permitting facile 18 F radionuclide incorporation required for PET imaging. The resulting molecule, [18 F]JW199, labels active NCEH1 in live cells at nanomolar concentrations and greater than 1000-fold selectivity relative to other serine hydrolases. [18 F]JW199 displays rapid, NCEH1-dependent accumulation in mouse tissues. Finally, we demonstrate that [18 F]JW199 labels aggressive cancer tumor cells in vivo, which uncovered localized NCEH1 activity at the leading edge of triple-negative breast cancer tumors, suggesting roles for NCEH1 in tumor aggressiveness and metastasis.


Subject(s)
Fluorine Radioisotopes/therapeutic use , Positron-Emission Tomography/methods , Sterol Esterase/metabolism , Animals , Female , Humans , Mice
7.
Article in English | MEDLINE | ID: mdl-27390783

ABSTRACT

Results from epidemiological studies suggest that there is an association between periodontitis and prediabetes, however, causality is not known. The results from our previous studies suggest that induction of periodontitis leads to hyperinsulinemia glucose intolerance and insulin resistance, all hallmarks of prediabetes. However, global effects of periodontitis on critical organs in terms of metabolic alterations are unknown. We determined the metabolic effects of periodontitis on brain, liver, heart and plasma resulting from Porphyromonas gingivalis induced periodontitis in mice. Periodontitis was induced by oral application of the periodontal pathogen, Porphyromonas gingivalis for 22 weeks. Global untargeted biochemical profiles in samples from these organs/plasma were determined by liquid and gas chromatography/mass spectrometry and compared between controls and animals with periodontitis. Oral application of Porphyromonas gingivalis induced chronic periodontitis and hallmarks of prediabetes. The results of sample analyses indicated a number of changes in metabolic readouts, including changes in metabolites related to glucose and arginine metabolism, inflammation and redox homeostasis. Changes in biochemicals suggested subtle systemic effects related to periodontal disease, with increases in markers of inflammation and oxidative stress most prominent in the liver. Signs of changes in redox homeostasis were also seen in the brain and heart. Elevated bile acids in liver were suggestive of increased biosynthesis, which may reflect changes in liver function. Interestingly, signs of decreasing glucose availability were seen in the brain. In all three organs and plasma, there was a significant increase in the microbiome-derived bioactive metabolite 4-ethylphenylsulfate sulfate in animals with periodontitis. The results of metabolic profiling suggest that periodontitis/bacterial products alter metabolomic signatures of brain, heart, liver, and plasma in the prediabetic state. These data provide scientific community valuable metabolic signatures that become the basis for understanding the impact of periodontitis on a systemic disease and potentially targets for therapeutic intervention.

8.
J Nucl Med ; 57(2): 279-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26564318

ABSTRACT

UNLABELLED: There is strong clinical interest in using neural stem cells (NSCs) as carriers for targeted delivery of therapeutics to glioblastoma. Multimodal dynamic in vivo imaging of NSC behaviors in the brain is necessary for developing such tailored therapies; however, such technology is lacking. Here we report a novel strategy for mesoporous silica nanoparticle (MSN)-facilitated NSC tracking in the brain via SPECT. METHODS: (111)In was conjugated to MSNs, taking advantage of the large surface area of their unique porous feature. A series of nanomaterial characterization assays was performed to assess the modified MSN. Loading efficiency and viability of NSCs with (111)In-MSN complex were optimized. Radiolabeled NSCs were administered to glioma-bearing mice via either intracranial or systemic injection. SPECT imaging and bioluminescence imaging were performed daily up to 48 h after NSC injection. Histology and immunocytochemistry were used to confirm the findings. RESULTS: (111)In-MSN complexes show minimal toxicity to NSCs and robust in vitro and in vivo stability. Phantom studies demonstrate feasibility of this platform for NSC imaging. Of significance, we discovered that decayed (111)In-MSN complexes exhibit strong fluorescent profiles in preloaded NSCs, allowing for ex vivo validation of the in vivo data. In vivo, SPECT visualizes actively migrating NSCs toward glioma xenografts in real time after both intracranial and systemic administrations. This is in agreement with bioluminescence live imaging, confocal microscopy, and histology. CONCLUSION: These advancements warrant further development and integration of this technology with MRI for multimodal noninvasive tracking of therapeutic NSCs toward various brain malignancies.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioblastoma/diagnostic imaging , Neural Stem Cells/diagnostic imaging , Tomography, Emission-Computed, Single-Photon/methods , Animals , Cell Line, Tumor , Humans , Indium Radioisotopes/adverse effects , Indium Radioisotopes/pharmacokinetics , Isotope Labeling , Magnetic Resonance Imaging , Mice , Mice, Nude , Multimodal Imaging , Nanoparticles , Phantoms, Imaging , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
9.
PLoS One ; 10(8): e0136502, 2015.
Article in English | MEDLINE | ID: mdl-26317345

ABSTRACT

BACKGROUND: Results from epidemiological studies indicate a close association between periodontitis and type 2 diabetes mellitus. However, the mechanism linking periodontitis to glucose intolerance (GI) and insulin resistance (IR) is unknown. We therefore tested the hypothesis that periodontitis induces the development of GI/IR through a liver Toll-like receptor 4 (TLR4) dependent mechanism. METHODS: TLR4 chimeric mice were developed by bone marrow transplantation using green fluorescent protein expressing TLR4WT mouse (GFPWT) as donor and TLR4 WT or TLR4-/- as recipient mice (GFPWT:WT and GFPWT:KO chimeras respectively). These chimeras were subjected to experimental chronic periodontitis induced by repeated applications of LPS to the gingival sulci for 18 weeks. The levels of GI/IR were monitored and plasma cytokines and LPS were determined at 18 weeks when differences in glucose tolerance were most apparent. Cytokine gene expression was measured in liver tissue by qPCR. RESULTS: Alveolar bone loss was significantly greater in GFPWT:WT chimeras treated with LPS compared with chimeras treated with PBS or GFPWT:KO chimeras. However, the degree of gingival inflammation was similar between GFPWT:WT and GFPWT:KO mice with LPS application. Severe GI/IR occurred in GFPWT:WT chimeras but not in the GFPWT:KO chimeras that were subjected to 18 weeks of LPS. Serum LPS was detected only in animals to which LPS was applied and the level was similar in GFPWT:WT and GFPWT:KO mice at the 18 week time point. Surprisingly, there was no significant difference in the plasma levels of IL1ß, IL6 and TNFα at 18 weeks in spite of the severe GI/IR in the GFPWT:WT chimeras with LPS application. Also, no difference in the expression of TNFα or IL6 mRNA was detected in the liver of GFPWT:WT vs GFPWT:KO mice. In contrast, liver IL1ß expression was significantly greater in GFPWT:WT chimeras compared to GFPWT:KO chimeras treated with LPS. CONCLUSION: We observed that GFPWT:WT, but not GFPWT:KO chimeras, treated with LPS developed GI/IR despite similar degrees of gingival inflammation, circulating cytokine levels, and LPS concentrations. We conclude that LPS from periodontitis sites has a pivotal role in triggering the development of GI/IR through a mechanism that involves TLR4 expression by resident macrophages/Kupffer cells in the liver.


Subject(s)
Gene Expression Regulation , Glucose Intolerance/metabolism , Insulin Resistance , Kupffer Cells/metabolism , Liver/metabolism , Periodontitis/metabolism , Toll-Like Receptor 4/biosynthesis , Allografts , Animals , Bone Marrow Transplantation , Chronic Disease , Disease Models, Animal , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Kupffer Cells/pathology , Lipopolysaccharides/toxicity , Liver/pathology , Mice , Mice, Knockout , Periodontitis/chemically induced , Periodontitis/genetics , Periodontitis/pathology , Toll-Like Receptor 4/genetics , Transplantation Chimera
10.
PLoS One ; 8(2): e57635, 2013.
Article in English | MEDLINE | ID: mdl-23451253

ABSTRACT

In this study we aim to boost the functional output of the intra-kidney islet transplantation for diabetic patients using a tissue engineered polymeric scaffold. This highly porous electrospun scaffold featured randomly distributed fibers composed of polycaprolactone (PCL) and poliglecaprone (PGC). It successfully sustained murine islets in vitro for up to 4 weeks without detected cytotoxicity. The in vivo study showed that the islet population proliferated by 89% within 12 weeks when they were delivered by the scaffold but only 18% if freely injected. Correspondingly, the islet population delivered by the scaffold unleashed a greater capability to produce insulin that in turn further drove down the blood glucose within 12 weeks after the surgery. Islets delivered by the scaffold most effectively prevented diabetic deterioration of kidney as evidenced by the lack of a kidney or glomerular enlargement and physiological levels of creatinine, urea nitrogen and albumin through week 12 after the surgery. Unlike traditional wisdom in diabetic research, the mechanistic study suggested that monocytes chemoattractant protein-1 (MCP-1) was responsible for the improved preservation of renal functions. This study revealed a therapeutic role of MCP-1 in rescuing kidneys in diabetic patients, which can be integrated into a tissue engineered scaffold to simultaneously preserved renal functions and islet transplantation efficacy. Also, this study affords a simple yet effective solution to improve the clinical output of islet transplantation.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL2/pharmacology , Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Kidney/surgery , Tissue Engineering/methods , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
Front Physiol ; 4: 11, 2013.
Article in English | MEDLINE | ID: mdl-23508205

ABSTRACT

Genetic variability has a profound effect on the development of cardiac hypertrophy in response to stress. Consequently, using a variety of inbred mouse strains with known genetic profiles may be powerful models for studying the response to cardiovascular stress. To explore this approach we looked at male C57BL/6J and 129/SvJ mice. Hemodynamic analyses of left ventricular pressures (LVPs) indicated significant differences in 129/SvJ and C57BL/6J mice that implied altered Ca(2+) handling. Specifically, 129/SvJ mice demonstrated reduced rates of relaxation and insensitivity to dobutamine (Db). We hypothesized that altered expression of genes controlling the influx and efflux of Ca(2+) from the sarcoplasmic reticulum (SR) was responsible and investigated the expression of several genes involved in maintaining the intracellular and sarcoluminal Ca(2+) concentration using quantitative real-time PCR analyses (qRT-PCR). We observed significant differences in baseline gene expression as well as different responses in expression to isoproterenol (ISO) challenge. In untreated control animals, 129/SvJ mice expressed 1.68× more ryanodine receptor 2(Ryr2) mRNA than C57BL/6J mice but only 0.37× as much calsequestrin 2 (Casq2). After treatment with ISO, sarco(endo)plasmic reticulum Ca(2+)-ATPase(Serca2) expression was reduced nearly two-fold in 129/SvJ while expression in C57BL/6J was stable. Interestingly, ß (1) adrenergic receptor(Adrb1) expression was lower in 129/SvJ compared to C57BL/6J at baseline and lower in both strains after treatment. Metabolically, the brain isoform of creatine kinase (Ckb) was up-regulated in response to ISO in C57BL/6J but not in 129/SvJ. These data suggest that the two strains of mice regulate Ca(2+) homeostasis via different mechanisms and may be useful in developing personalized therapies in human patients.

12.
Magn Reson Imaging ; 31(4): 508-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23102946

ABSTRACT

Autoimmune ablation of pancreatic ß-cells and alteration of its microvasculature may be a predictor of Type I diabetes development. A dynamic manganese-enhanced MRI (MEMRI) approach and an empirical mathematical model were developed to monitor whole pancreatic ß-cell function and vasculature modifications in mice. Normal and streptozotocin-induced diabetic FVB/N mice were imaged on a 9.4T MRI system using a 3D magnetization prepared rapid acquisition gradient echo pulse sequence to characterize low dose manganese kinetics in the pancreas head, body and tail. Average signal enhancement in the pancreas (head, body, and tail) as a function of time was fit by a novel empirical mathematical model characterizing contrast uptake/washout rates and yielding parameters describing peak signal, initial slope, and initial area under the curve. Signal enhancement from glucose-induced manganese uptake was fit by a linear function. The results demonstrated that the diabetic pancreatic tail had a significantly lower contrast uptake rate, smaller initial slope/initial area under the curve, and a smaller rate of Mn uptake following glucose activation (p<0.05) compared to the normal pancreatic tail. These observations parallel known patterns of ß-cell loss and alteration in supportive vasculature associated with diabetes. Dynamic MEMRI is a promising technique for assessing ß-cell functionality and vascular perfusion with potential applications for monitoring diabetes progression and/or therapy.


Subject(s)
Chlorides/pharmacokinetics , Glucose/pharmacokinetics , Image Interpretation, Computer-Assisted/methods , Insulin-Secreting Cells/metabolism , Magnetic Resonance Imaging/methods , Manganese Compounds/pharmacokinetics , Models, Biological , Animals , Computer Simulation , Contrast Media/pharmacokinetics , Diabetes Mellitus, Experimental/chemically induced , Male , Metabolic Clearance Rate , Mice , Streptozocin
13.
Contrast Media Mol Imaging ; 6(6): 474-81, 2011.
Article in English | MEDLINE | ID: mdl-22144025

ABSTRACT

Manganese (Mn) is a calcium (Ca) analog that has long been used as a magnetic resonance imaging (MRI) contrast agent for investigating cardiac tissue functionality, for brain mapping and for neuronal tract tracing studies. Recently, we have extended its use to investigate pancreatic ß-cells and showed that, in the presence of MnCl(2), glucose-activated pancreatic islets yield significant signal enhancement in T(1)-weigheted MR images. In this study, we exploited for the first time the unique capabilities of X-ray fluorescence microscopy (XFM) to both visualize and quantify the metal in pancreatic ß-cells at cellular and subcellular levels. MIN-6 insulinoma cells grown in standard tissue culture conditions had only a trace amount of Mn, 1.14 ± 0.03 × 10(-11)µg/µm(2), homogenously distributed across the cell. Exposure to 2 mM glucose and 50 µM MnCl(2) for 20 min resulted in nonglucose-dependent Mn uptake and the overall cell concentration increased to 8.99 ± 2.69 × 10(-11) µg/µm(2). When cells were activated by incubation in 16 mM glucose in the presence of 50 µM MnCl(2), a significant increase in cytoplasmic Mn was measured, reaching 2.57 ± 1.34 × 10(-10) µg/µm(2). A further rise in intracellular concentration was measured following KCl-induced depolarization, with concentrations totaling 1.25 ± 0.33 × 10(-9) and 4.02 ± 0.71 × 10(-10) µg/µm(2) in the cytoplasm and nuclei, respectively. In both activated conditions Mn was prevalent in the cytoplasm and localized primarily in a perinuclear region, possibly corresponding to the Golgi apparatus and involving the secretory pathway. These data are consistent with our previous MRI findings, confirming that Mn can be used as a functional imaging reporter of pancreatic ß-cell activation and also provide a basis for understanding how subcellular localization of Mn will impact MRI contrast.


Subject(s)
Chlorides/pharmacokinetics , Contrast Media/pharmacokinetics , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Magnetic Resonance Imaging/methods , Manganese Compounds/pharmacokinetics , Microscopy, Fluorescence/methods , Cell Line, Tumor , Humans , Insulin-Secreting Cells/drug effects , Pancreatic Neoplasms/diagnosis , X-Rays
14.
NMR Biomed ; 23(10): 1158-65, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21162143

ABSTRACT

The noninvasive assessment of pancreatic islets would be an invaluable tool in advancing the treatment of type I diabetes and in understanding its pathophysiology. As shown previously in rodents, manganese-enhanced MRI (MEMRI) can be successfully used to quantify ß-cell function. In this study, we successfully applied this technique to isolated human pancreatic islets in both a static and, more significantly, MRI-compatible perfusion set-up. Unlike rodent islets, which produced a significant increase in the signal-to-noise ratio (SNR) when treated with 25 µM MnCl(2) or less, human islets demonstrated significant manganese uptake when exposed to an extracellular concentration of 50 µM MnCl(2). Nonspecific passive manganese uptake was present and quantified in a 15% SNR increase over the control group. However, glucose-induced manganese uptake caused an SNR increase equal to 45% over nonactivated islets. This corresponds to a statistically significant decrease in the T(1) relaxation time from 1501 ms for untreated islets to 1362 ms following passive uptake, and to 861 ms following glucose stimulation. As expected, no manganese cytotoxicity was measured, as shown by normal insulin secretion profiles. These data confirm the viability of MEMRI to assess isolated human islet functionality in vitro, and this technique shows promise for the monitoring of their performance in vivo following transplantation.


Subject(s)
Islets of Langerhans/anatomy & histology , Magnetic Resonance Imaging/methods , Tissue Culture Techniques/methods , Humans , Image Enhancement , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Manganese/pharmacology , Perfusion , Signal Processing, Computer-Assisted , Spin Labels
15.
Biomaterials ; 28(21): 3188-97, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17449092

ABSTRACT

A goal of current orthopedic biomaterials research is to design implants that induce controlled, guided, and rapid healing. In addition to acceleration of normal wound healing phenomena, these implants should result in the formation of a characteristic interfacial layer with adequate biomechanical properties. To achieve these goals, however, a better understanding of events at the bone-material interface is needed, as well as the development of new materials and approaches that promote osseointegration. Using anodization, titania interfaces can be fabricated with controlled nanoarchitecture. This study demonstrates the ability of these surfaces to promote osteoblast differentiation and matrix production, and enhance short- and long-term osseointegration in vitro. Titania nanotubular surfaces were fabricated using an anodization technique. Marrow stromal cells (MSCs) were isolated from male Lewis rats and seeded on these surfaces along with control surfaces. The interaction of cells with these surfaces was investigated in terms of their ability to adhere, proliferate and differentiate on them. The experiments were repeated three times with cells from different cultures. All the results were analyzed using analysis of variance (ANOVA). Statistical significance was considered at p<0.05. Furthermore, in vivo biocompatibility was assessed by implanting surfaces subcutaneously in male Lewis rat and performing histological analysis after 4 weeks. Our results indicate that the nanotubular titania surfaces provide a favorable template for the growth and maintenance of bone cells. The cells cultured on nanotubular surfaces showed higher adhesion, proliferation, ALP activity and bone matrix deposition compared to those grown on flat titanium surfaces. In vivo biocompatibility results suggest that nanotubular titania does not cause chronic inflammation or fibrosis. The fabrication routes of titania nano-architectures are flexible and cost-effective, enabling realization of desired platform topologies on existing non-planar orthopedic implants.


Subject(s)
Bone Marrow Cells/cytology , Engineering , Nanotechnology/methods , Nanotubes/chemistry , Titanium/chemistry , Alkaline Phosphatase/analysis , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials/chemistry , Bone Marrow Cells/ultrastructure , Bone and Bones/cytology , Calcium/analysis , Calcium/metabolism , Cell Adhesion , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Fluoresceins/metabolism , Formazans/metabolism , Hot Temperature , Male , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteoblasts/physiology , Osteoblasts/ultrastructure , Rats , Rats, Inbred Lew , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/physiology , Stromal Cells/ultrastructure , Tetrazolium Salts/metabolism , Time Factors
16.
Biomaterials ; 28(16): 2638-45, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17335895

ABSTRACT

Cellular immunoisolation using semi-permeable barriers has been investigated over the past several decades as a promising treatment approach for diseases such as Parkinson's, Alzheimer's, and Type 1 diabetes. Typically, polymeric membranes are used for immunoisolation applications; however, recent advances in technology have led to the development of more robust membranes that are able to more completely meet the requirements for a successful immunoisolation device, including well controlled pore size, chemical and mechanical stability, nonbiodegradability, and biocompatibility with both the graft tissue as well as the host. It has been shown previously that nanoporous alumina biocapsules can act effectively as immunoisolation devices, and support the viability and functionality of encapsulated beta cells. The aim of this investigation was to assess the biocompatibility of the material with host tissue. The cytotoxicity of the capsule, as well as its ability to activate complement and inflammation was studied. Further, the effects of poly(ethylene glycol) (PEG) modification on the tissue response to implanted capsules were studied. Our results have shown that the device is nontoxic and does not induce significant complement activation. Further, in vivo work has demonstrated that implantation of these capsules into the peritoneal cavity of rats induces a transient inflammatory response, and that PEG is useful in minimizing the host response to the material.


Subject(s)
Aluminum Oxide , Coated Materials, Biocompatible/metabolism , Immunologic Techniques/instrumentation , Membranes, Artificial , Nanostructures , Aluminum Oxide/chemistry , Aluminum Oxide/metabolism , Animals , Capsules/chemistry , Capsules/metabolism , Coated Materials, Biocompatible/chemistry , Complement Activation , Materials Testing , Polyethylene Glycols/chemistry , Rats , Surface Properties
17.
Cell Transplant ; 15(2): 195-203, 2006.
Article in English | MEDLINE | ID: mdl-16719054

ABSTRACT

The increasing incidence of diabetes and the need to further understand its cellular basis has resulted in the development of new diagnostic and therapeutic techniques. Nonetheless, the quest to noninvasively ascertain beta-cell mass and function has not been achieved. Manganese (Mn)-enhanced MRI is presented here as a tool to image beta-cell functionality in cell culture and isolated islets. Similar to calcium, extracellular Mn was taken up by glucose-activated beta-cells resulting in 200% increase in MRI contrast enhancement, versus nonactivated cells. Similarly, glucose-activated islets showed an increase in MRI contrast up to 45%. Although glucose-stimulated Ca influx was depressed in the presence of 100 microM Mn, no significant effect was seen at lower Mn concentrations. Moreover, islets exposed to Mn showed normal glucose sensitivity and insulin secretion. These results demonstrate a link between image contrast enhancement and beta-cell activation in vitro, and provide the basis for future noninvasive in vivo imaging of islet functionality and beta-cell mass.


Subject(s)
Image Enhancement/methods , Insulin-Secreting Cells/metabolism , Magnetic Resonance Imaging/methods , Animals , Cell Line, Tumor , Cell Separation , Cells, Cultured , Dose-Response Relationship, Drug , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/chemistry , Insulinoma/chemistry , Insulinoma/metabolism , Insulinoma/physiopathology , Islets of Langerhans Transplantation/methods , Male , Manganese/pharmacokinetics , Manganese/pharmacology , Mice , Mice, Inbred Strains , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/physiopathology , Rats , Rats, Inbred Lew
18.
Adv Drug Deliv Rev ; 56(2): 211-29, 2004 Feb 10.
Article in English | MEDLINE | ID: mdl-14741117

ABSTRACT

In recent years, rapid advancements have been made in the biomedical applications of micro- and nanotechnology. While the focus of such technology has primarily been on in vitro analytical and diagnostic tools, more recently, in vivo therapeutic and sensing applications have gained attention. The long-term integration of cells with inorganic materials provides the basis for novel delivery and sensing platforms. Our recent work has focused on the ability to maintain cells long term in nanoporous silicon-based microenvironments. This paper describes the creation of monodisperse, nanoporous, biocompatible, silicon membranes as a platform for the delivery of cells. Studies described herein focus on the interaction of silicon-based substrates with cells of interest in terms of viability, proliferation, and functionality. Such microfabricated nanoporous membranes can be used both in vitro for cell-based assays and in vivo for immunoisolation and drug delivery applications.


Subject(s)
Capsules , Drug Delivery Systems/instrumentation , Animals , Biocompatible Materials , Drug Delivery Systems/methods , Equipment Design , Glucose/chemistry , Insulin/administration & dosage , Insulin/chemistry , Membranes, Artificial , Permeability , Silicones
SELECTION OF CITATIONS
SEARCH DETAIL
...