Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(3): e0299961, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38483851

RESUMO

In vivo noninvasive imaging of neurometabolites is crucial to improve our understanding of the underlying pathophysiological mechanism in neurodegenerative diseases. Abnormal changes in synaptic organization leading to synaptic degradation and neuronal loss is considered as one of the primary factors driving Alzheimer's disease pathology. Magnetic resonance based molecular imaging techniques such as chemical exchange saturation transfer (CEST) and magnetic resonance spectroscopy (MRS) can provide neurometabolite specific information which may relate to underlying pathological and compensatory mechanisms. In this study, CEST and short echo time single voxel MRS was performed to evaluate the sensitivity of cerebral metabolites to beta-amyloid (Aß) induced synaptic deficit in the hippocampus of a mouse model of Alzheimer's disease. The CEST based spectra (Z-spectra) were acquired on a 9.4 Tesla small animal MR imaging system with two radiofrequency (RF) saturation amplitudes (1.47 µT and 5.9 µT) to obtain creatine-weighted and glutamate-weighted CEST contrasts, respectively. Multi-pool Lorentzian fitting and quantitative T1 longitudinal relaxation maps were used to obtain metabolic specific apparent exchange-dependent relaxation (AREX) maps. Short echo time (TE = 12 ms) single voxel MRS was acquired to quantify multiple neurometabolites from the right hippocampus region. AREX contrasts and MRS based metabolite concentration levels were examined in the ARTE10 animal model for Alzheimer's disease and their wild type (WT) littermate counterparts (age = 10 months). Using MRS voxel as a region of interest, group-wise analysis showed significant reduction in Glu-AREX and Cr-AREX in ARTE10, compared to WT animals. The MRS based results in the ARTE10 mice showed significant decrease in glutamate (Glu) and glutamate-total creatine (Glu/tCr) ratio, compared to WT animals. The MRS results also showed significant increase in total creatine (tCr), phosphocreatine (PCr) and glutathione (GSH) concentration levels in ARTE10, compared to WT animals. In the same ROI, Glu-AREX and Cr-AREX demonstrated positive associations with Glu/tCr ratio. These results indicate the involvement of neurotransmitter metabolites and energy metabolism in Aß-mediated synaptic degradation in the hippocampus region. The study also highlights the feasibility of CEST and MRS to identify and track multiple competing and compensatory mechanisms involved in heterogeneous pathophysiology of Alzheimer's disease in vivo.


Assuntos
Doença de Alzheimer , Creatina , Camundongos , Animais , Creatina/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Animais Selvagens/metabolismo , Ácido Glutâmico , Receptores de Antígenos de Linfócitos T
2.
Front Oncol ; 12: 939260, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36483050

RESUMO

Many drugs that show potential in animal models of glioblastoma (GBM) fail to translate to the clinic, contributing to a paucity of new therapeutic options. In addition, animal model development often includes histologic assessment, but multiparametric/multimodality imaging is rarely included despite increasing utilization in patient cancer management. This study developed an intracranial recurrent, drug-resistant, human-derived glioblastoma tumor in Sprague-Dawley Rag2-Rag2 tm1Hera knockout rat and was characterized both histologically and using multiparametric/multimodality neuroimaging. Hybrid 18F-fluoroethyltyrosine positron emission tomography and magnetic resonance imaging, including chemical exchange saturation transfer (18F-FET PET/CEST MRI), was performed for full tumor viability determination and characterization. Histological analysis demonstrated human-like GBM features of the intracranially implanted tumor, with rapid tumor cell proliferation (Ki67 positivity: 30.5 ± 7.8%) and neovascular heterogeneity (von Willebrand factor VIII:1.8 to 5.0% positivity). Early serial MRI followed by simultaneous 18F-FET PET/CEST MRI demonstrated consistent, predictable tumor growth, with exponential tumor growth most evident between days 35 and 49 post-implantation. In a second, larger cohort of rats, 18F-FET PET/CEST MRI was performed in mature tumors (day 49 post-implantation) for biomarker determination, followed by evaluation of single and combination therapy as part of the model development and validation. The mean percentage of the injected dose per mL of 18F-FET PET correlated with the mean %CEST (r = 0.67, P < 0.05), but there was also a qualitative difference in hot spot location within the tumor, indicating complementary information regarding the tumor cell demand for amino acids and tumor intracellular mobile phase protein levels. Finally, the use of this glioblastoma animal model for therapy assessment was validated by its increased overall survival after treatment with combination therapy (temozolomide and idasanutlin) (P < 0.001). Our findings hold promise for a more accurate tumor viability determination and novel therapy assessment in vivo in a recently developed, reproducible, intracranial, PDX GBM.

3.
Pediatr Blood Cancer ; 67(2): e28081, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31724812

RESUMO

BACKGROUND: Neuroblastoma, the most common extracranial solid tumor of childhood, produces catecholamines that are metabolized within tumor cells. Homovanillic acid (HVA) and vanillylmandelic acid (VMA), the end products of catecholamine metabolism, have limited accuracy for testing of the tumors. This study assessed whether metabolites produced in earlier steps of catecholamine metabolism might offer improved diagnostic accuracy over urinary HVA and VMA. PROCEDURE: Plasma concentrations of 3-methoxytyramine, normetanephrine, and metanephrine were measured in two pediatric cohorts: (i) 96 children with confirmed neuroblastoma and (ii) 41 children with signs and symptoms of a catecholamine-producing tumor or other neoplasms and in whom neuroblastoma was excluded. Additional measurements of plasma 3-O-methyldopa and relationships of metabolites to MYCN amplification were examined in patient subgroups. RESULTS: Overall, 94 of the 96 patients with neuroblastoma had concentrations of 3-methoxytyramine or normetanephrine above age-specific upper limits of reference intervals, providing a diagnostic sensitivity of 97.9% that was higher (P < 0.0001) than that of 82.2% for HVA and VMA. One of the two patients with normal plasma results showed an elevation of plasma 3-O-methyldopa. Diagnostic specificities were, respectively, 95.1% and 84.8%. Areas under receiver-operating characteristic curves confirmed the superior diagnostic power of the plasma than the urinary test (0.994 vs 0.945; P = 0.0095). Ratios of plasma 3-methoxytyramine to normetanephrine were 7.2-fold higher (P < 0.0001) for patients who had neuroblastomas with MYCN amplification than without MYCN amplification. CONCLUSIONS: Measurements of plasma 3-methoxytyramine and normetanephrine provide a highly accurate diagnostic test for neuroblastoma and also offer potential for prognostic risk stratification.


Assuntos
Biomarcadores Tumorais/análise , Dopamina/análogos & derivados , Neuroblastoma/diagnóstico , Normetanefrina/análise , Tirosina/análogos & derivados , Adolescente , Estudos de Casos e Controles , Criança , Pré-Escolar , Dopamina/análise , Feminino , Seguimentos , Humanos , Lactente , Masculino , Neuroblastoma/sangue , Neuroblastoma/urina , Prognóstico , Estudos Retrospectivos , Tirosina/análise
4.
Cancer Res ; 79(12): 3112-3124, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31015228

RESUMO

The cell membrane glycolipid GD2 is expressed by multiple solid tumors, including 88% of osteosarcomas and 98% of neuroblastomas. However, osteosarcomas are highly heterogeneous, with many tumors exhibiting GD2 expression on <50% of the individual cells, while some tumors are essentially GD2-negative. Anti-GD2 immunotherapy is the current standard of care for high-risk neuroblastoma, but its application to recurrent osteosarcomas, for which no effective therapies exist, has been extremely limited. This is, in part, because the standard assays to measure GD2 expression in these heterogeneous tumors are not quantitative and are subject to tissue availability and sampling bias. To address these limitations, we evaluated a novel, sensitive radiotracer [64Cu]Cu-Bn-NOTA-hu14.18K322A to detect GD2 expression in osteosarcomas (six patient-derived xenografts and one cell line) in vivo using positron emission tomography (PET). Tumor uptake of the radiolabeled, humanized anti-GD2 antibody [64Cu]Cu-Bn-NOTA-hu14.18K322A was 7-fold higher in modestly GD2-expressing osteosarcomas (32% GD2-positive cells) than in a GD2-negative tumor (9.8% vs. 1.3% of the injected dose per cc, respectively). This radiotracer also identified lesions as small as 29 mm3 in a 34% GD2-positive model of metastatic osteosarcoma of the lung. Radiolabeled antibody accumulation in patient-derived xenografts correlated with GD2 expression as measured by flow cytometry (Pearson r = 0.88, P = 0.01), distinguishing moderately GD2-expressing osteosarcomas (32%-69% GD2-positive cells) from high GD2 expressors (>99%, P < 0.05). These results support the utility of GD2 imaging with PET to measure GD2 expression in osteosarcoma and thus maximize the clinical impact of anti-GD2 immunotherapy. SIGNIFICANCE: In situ assessment of all GD2-positive osteosarcoma sites with a novel PET radiotracer could significantly impact anti-GD2 immunotherapy patient selection and enable noninvasive probing of correlations between target expression and therapeutic response.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Ósseas/patologia , Gangliosídeos/antagonistas & inibidores , Neoplasias Pulmonares/secundário , Recidiva Local de Neoplasia/patologia , Osteossarcoma/patologia , Tomografia por Emissão de Pósitrons/métodos , Animais , Apoptose , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/metabolismo , Proliferação de Células , Gangliosídeos/imunologia , Humanos , Imunoterapia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/metabolismo , Osteossarcoma/tratamento farmacológico , Osteossarcoma/imunologia , Osteossarcoma/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Labelled Comp Radiopharm ; 61(14): 1069-1080, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30289176

RESUMO

6-[18 F]Fluorodopamine ([18 F]F-DA) is taken into cells via the norepinephrine transporter (NET). Recent [18 F]F-DA positron emission tomography-computed tomography (PET-CT) imaging of adult neuroendocrine tumors shows a dramatic improvement in sensitivity over the standard-of-care, meta-iodobenzylguanidine (MIBG) single-photon emission computed tomography (SPECT)-CT. A new precursor (ALPdopamine™) allows no-carrier-added synthesis resulting in high-molar activity [18 F]F-DA. Automated synthesis of [18 F]F-DA was performed in a single reactor using a two-step procedure: 1) fluorination via thermolysis of a diaryliodonium salt precursor, followed by 2) acid hydrolysis. Phase transfer agents, Kryptofix 222 and two tetraalkylammonium salts, were investigated. Optimized synthesis of [18 F]F-DA was achieved in 56 to 60 minutes (26% end of synthesis [EOS], nondecay corrected). The product passed all Food and Drug Administration (FDA)-required quality control testing for human use. Accumulation of [18 F]F-DA in SK-N-BE(2)-C (high NET expression) cells was significantly higher than in SH-EP (minimal NET expression) cells (P < 0.0001). ALPdopamine provides an effective scaffold for the routine production of [18 F]F-DA for human use. Validation of uptake by neuroblastoma (NB) cell lines supports the use of [18 F]F-DA for imaging NB patients. A pediatric NB imaging trial using [18 F]F-DA PET has been approved (Investigational New Drug application (IND) no. 138638) based on the methods reported here. We expect [18 F]F-DA will be localized in NB tumors and that high-quality functional images will be obtained within minutes after injection.


Assuntos
Dopamina/análogos & derivados , Neuroblastoma/diagnóstico por imagem , Transporte Biológico , Linhagem Celular Tumoral , Técnicas de Química Sintética , Dopamina/síntese química , Dopamina/química , Dopamina/metabolismo , Humanos , Neuroblastoma/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Controle de Qualidade , Radioquímica
6.
Theranostics ; 8(10): 2722-2738, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29774071

RESUMO

Exosomes are a potential source of cancer biomarkers. Probing tumor-derived exosomes can offer a potential non-invasive way to diagnose cancer, assess cancer progression, and monitor treatment responses. Novel molecular methods would facilitate exosome analysis and accelerate basic and clinical exosome research. Methods: A standard gold-coated glass microscopy slide was used to develop a miniaturized affinity-based device to capture exosomes in a target-specific manner with the assistance of low-cost 3-D printing technology. Gold nanorods coated with QSY21 Raman reporters were used as the label agent to quantitatively detect the target proteins based on surface enhanced Raman scattering spectroscopy. The expressions of several surface protein markers on exosomes from conditioned culture media of breast cancer cells and from HER2-positive breast cancer patients were quantitatively measured. The data was statistically analyzed and compared with healthy controls. Results: A miniaturized 17 × 5 Au array device with 2-mm well size was fabricated to capture exosomes in a target-specific manner and detect the target proteins on exosomes with surface enhanced Raman scattering gold nanorods. This assay can specifically detect exosomes with a limit of detection of 2×106 exosomes/mL and analyze over 80 purified samples on a single device within 2 h. Using the assay, we have showed that exosomes derived from MDA-MB-231, MDA-MB-468, and SKBR3 breast cancer cells give distinct protein profiles compared to exosomes derived from MCF12A normal breast cells. We have also showed that exosomes in the plasma from HER2-positive breast cancer patients exhibit significantly (P ≤ 0.01) higher level of HER2 and EpCAM than those from healthy donors. Conclusion: We have developed a simple, inexpensive, highly efficient, and portable Raman exosome assay for detection and protein profiling of exosomes. Using the assay and model exosomes from breast cancer cells, we have showed that exosomes exhibit diagnostic surface protein markers, reflecting the protein profile of their donor cells. Through proof-of-concept studies, we have identified HER2 and EpCAM biomarkers on exosomes in plasma from HER2-positive breast cancer patients, suggesting the diagnostic potential of these markers for breast cancer diagnostics. This assay would accelerate exosome research and pave a way to the development of novel cancer liquid biopsy for cancer detection and monitoring.


Assuntos
Neoplasias da Mama/sangue , Exossomos/metabolismo , Nanotubos/química , Microscopia Óptica não Linear/instrumentação , Nanomedicina Teranóstica/instrumentação , Biomarcadores Tumorais/sangue , Linhagem Celular Tumoral , Feminino , Ouro/química , Humanos , Microscopia Óptica não Linear/métodos , Impressão Tridimensional/instrumentação , Receptor ErbB-2/metabolismo , Nanomedicina Teranóstica/métodos
7.
Clin Nucl Med ; 42(5): e275-e276, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28319499

RESUMO

Elevated relative cerebral blood volume on perfusion MRI and increased uptake on C-methionine PET can be used to diagnose and guide biopsy of brain tumors but are not specific. We report increased uptake on C-methionine PET associated with 4 developmental venous anomalies (DVAs) in 3 children with brain tumors, which could potentially mimic tumor and misdirect biopsy. Because DVAs are not readily visible on CT, prevention of misdirected biopsy in patients with focally elevated C-methionine uptake and relative cerebral blood volume relies on close correlation with contrast-enhanced anatomic MRI to exclude DVA or other nonneoplastic etiology.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Veias Cerebrais/anormalidades , Tomografia por Emissão de Pósitrons , Encéfalo/diagnóstico por imagem , Neoplasias Encefálicas/irrigação sanguínea , Veias Cerebrais/diagnóstico por imagem , Criança , Diagnóstico Diferencial , Humanos , Imageamento por Ressonância Magnética/métodos , Metionina , Pessoa de Meia-Idade , Perfusão
8.
J Mater Chem B ; 4(3): 513-520, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-27087966

RESUMO

Although nanomaterials have been widely investigated for drug delivery, imaging and immunotherapy, their potential roles in triggering innate cellular immune responses while simultaneously serving as imaging enhancer remain unexplored. In this work, gold nanoparticles (GNPs) conjugated to the tumor-targeting anti-GD2 antibody hu14.18K322A, namely HGNPs, were designed and synthesized to specifically enhance computerized tomography (CT) imaging contrast and to stimulate the attack of neuroblastoma and melanoma cells by natural killer (NK) cells. The HGNPs specifically targeted GD2-positive neuroblastoma (NB1691) and melanoma (M21) cells, with an enhancement of CT contrast images of the HGNP-labeled cell pellets by 5.27- and 7.66-fold, respectively, compared to images of unlabeled cell pellets. The HGNPs also triggered NK-mediated antibody-dependent cellular cytotoxicity (ADCC) in NB1691 and M21 cells with a two-fold higher efficacy compared to that elicited by hu14.18K322A alone, with no adverse effect to GD2-negative PC-3 cells. These results suggest that HGNPs are promising theranostic agents for neuroblastoma and melanoma cancers.

9.
J Nucl Med ; 54(11): 1902-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24050936

RESUMO

UNLABELLED: The purpose of this study was to evaluate the biodistribution of (11)C-labeled methionine in non-tumor-involved organs in pediatric patients studied for malignant diseases. METHODS: Ninety-three children and young adults with known or suspected malignancies underwent (11)C-methionine PET and CT scans. Imaging began 5-15 min after injection of 740 MBq (20 mCi) per 1.7 m(2) of body surface area. Images were acquired from the top of the head through the mid thighs. Standardized uptake values were determined using regions of interest drawn on the CT image and transferred to the corresponding transverse PET slice. RESULTS: The highest concentrations of (11)C-methionine were found in the pancreas and liver. Less intense uptake was seen in other regions, such as the salivary glands, tonsils, and bone marrow. There was little uptake in the lungs, fat (including brown adipose tissue), and muscle. Uptake in bone marrow, parotid glands, and tonsils was slightly but statistically significantly higher in men than women. Testicular, bone marrow, and left ventricular uptake increased with age. There was little variability statistically between comparisons of uptake change and groupings of age, race, sex, and patients studied at the time of diagnosis versus previously treated patients. CONCLUSION: High uptake of (11)C-methionine is reliably found in the pancreas and liver, consistent with the anabolic functions of these organs. Low uptake in the brain, neck, chest, pelvis, and extremities will facilitate tumor localization in those areas. However, intense uptake in the upper abdomen may limit the diagnostic utility of (11)C-methionine in that area.


Assuntos
Metionina/farmacocinética , Adolescente , Adulto , Fatores Etários , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Tomografia por Emissão de Pósitrons , Radiometria , Recidiva , Fatores Sexuais , Distribuição Tecidual , Tomografia Computadorizada por Raios X , Adulto Jovem
10.
J Nucl Med ; 53(11): 1772-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23064212

RESUMO

UNLABELLED: The hu14.18K322A variant of the GD2-targeting antibody hu14.18 has been shown to elicit a level of antibody-dependent cell-mediated cytotoxicity toward human neuroblastoma cells similar to that of the parent antibody. However, hu14.18K322A exhibited a decreased complement activation and associated pain, the dose-limiting toxicity in neuroblastoma immunotherapy. PET with a radiolabeled analog of the same antibody used in treatment will provide insight into the ability of hu14.18K322A to reach its target, as well as nontarget uptake that may cause side effects. Such antibody radiotracers might also provide a method for measuring GD2 expression in tumors, thus enabling the prediction of response to anti-GD2 therapy for individual patients. METHODS: The conjugation of hu14.18K322A with p-NH(2)-Bn-DOTA was accomplished using N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide with subsequent (64)Cu radiolabeling at 37°C for 30 min. Immunoreactivity of the conjugate was assessed by a dose-escalation blocking experiment measuring binding to purified GD2 versus GD1b as a negative control. Cell uptake and biodistribution studies in M21 (GD2-positive) and PC-3 (GD2-negative) tumor models were performed, as was small-animal PET/CT of M21 and PC-3 tumor-bearing mice. RESULTS: The labeling of (64)Cu-p-NH(2)-Bn-DOTA-hu14.18K322A was achieved at more than 95% radiochemical purity and a specific activity of 127-370 MBq/mg (3.4-10 mCi/mg) after chromatographic purification. Preliminary in vitro data demonstrated a greater than 6-fold selectivity of binding to GD2 versus GD1b and dose-dependent inhibition of binding by unmodified hu14.8K322A. In vivo data, including small-animal PET/CT, showed significant GD2-positive tumor-targeting ability, with a persistent 2-fold-higher uptake of radiotracer than in GD2-negative tumors. CONCLUSION: (64)Cu-p-NH(2)-Bn-DOTA-hu14.18K322A represents a novel PET radiotracer to facilitate clinical investigations of anti-GD2 immunotherapies and to complement other imaging modalities in the staging and treatment of neuroblastoma.


Assuntos
Anticorpos , Radioisótopos de Cobre , Compostos Heterocíclicos com 1 Anel/química , Melanoma/diagnóstico por imagem , Neuroblastoma/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Proteínas Recombinantes de Fusão , Animais , Anticorpos/química , Anticorpos/imunologia , Anticorpos/metabolismo , Linhagem Celular Tumoral , Feminino , Gangliosídeos/imunologia , Meia-Vida , Humanos , Melanoma/patologia , Camundongos , Neuroblastoma/patologia , Traçadores Radioativos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética
11.
J Am Chem Soc ; 133(35): 13918-21, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21827210

RESUMO

To conquer complex and devastating diseases such as cancer, more coordinated and combined attack strategies are needed. We suggest that these can be beautifully achieved by using nanoconstruct design. We present an example showing that neuroblastoma cells are selectively killed by a nanoconstruct that specifically targets neuroblastoma cells, pushes cells to the vulnerable phase of the cell cycle, and greatly enhances radiation-induced cell death. The success of this multipronged attack approach launched by cell-embedded nanoconstructs demonstrates the power and flexibility of nanotechnology in treating cancer, a difficult task for a small molecule.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Nanoestruturas/química , Neuroblastoma/terapia , Paclitaxel/administração & dosagem , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Nanomedicina/métodos , Nanoestruturas/análise , Raios X
12.
ACS Nano ; 5(6): 4581-91, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21595480

RESUMO

Carbon nanotubes (CNTs) cause perturbations in immune systems and limit the application of CNTs in biomedicine. Here we demonstrate that a surface chemistry modification on multiwalled CNTs (MWCNTs) reduces their immune perturbations in mice and in macrophages. The modified MWCNTs change their preferred binding pattern from mannose receptor to scavenger receptor. This switch significantly alleviates NFκB activation and reduces immunotoxicity of MWCNTs.


Assuntos
Nanotubos de Carbono/química , Animais , Inflamação , Interleucina-1beta/metabolismo , Lectinas Tipo C/química , Lipopolissacarídeos/química , Macrófagos/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/química , Camundongos , NF-kappa B/química , Nanoestruturas/química , Nanotecnologia/métodos , Ligação Proteica , Receptores de Superfície Celular/química , Receptores Depuradores/química , Propriedades de Superfície , Fator de Necrose Tumoral alfa/metabolismo
13.
Nat Nanotechnol ; 5(9): 683-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20693989

RESUMO

Soluble carbon nanotubes show promise as materials for in vivo delivery and imaging applications. Several reports have described the in vivo toxicity of carbon nanotubes, but their effects on male reproduction have not been examined. Here, we show that repeated intravenous injections of water-soluble multiwalled carbon nanotubes into male mice can cause reversible testis damage without affecting fertility. Nanotubes accumulated in the testes, generated oxidative stress and decreased the thickness of the seminiferous epithelium in the testis at day 15, but the damage was repaired at 60 and 90 days. The quantity, quality and integrity of the sperm and the levels of three major sex hormones were not significantly affected throughout the 90-day period. The fertility of treated male mice was unaffected; the pregnancy rate and delivery success of female mice that mated with the treated male mice did not differ from those that mated with untreated male mice.


Assuntos
Nanotubos de Carbono/efeitos adversos , Testículo/patologia , Animais , Feminino , Fertilidade , Hormônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanotubos de Carbono/química , Estresse Oxidativo , Análise do Sêmen , Testículo/ultraestrutura
14.
Nucl Med Biol ; 37(1): 3-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20122661

RESUMO

INTRODUCTION: The sensitivity of the in vivo binding of [(11)C]dihydrotetrabenazine ([(11)C]DTBZ) and [(11)C]methylphenidate ([(11)C]MPH) to their respective targets - vesicular monoamine transporter type 2 (VMAT2) and neuronal membrane dopamine transporter - after alterations in endogenous levels of dopamine was examined in the rat brain. METHODS: In vivo binding of [(11)C]DTBZ and [(11)C]MPH was determined using a bolus+infusion protocol. The in vitro number of VMAT2 binding sites was determined by autoradiography. RESULTS: Repeated dosing with alpha-methyl-p-tyrosine (AMPT) at doses that significantly (-75%) depleted brain tissue dopamine levels resulted in increased (+36%) in vivo [(11)C]DTBZ binding to VMAT2 in the striatum. The increase in binding could be completely reversed via treatment with L-DOPA/benserazide to restore dopamine levels. There were no changes in the total number of VMAT2 binding sites, as measured using in vitro autoradiography. No changes were observed for in vivo [(11)C]MPH binding to the dopamine transporter in the striatum following AMPT pretreatment. CONCLUSION: These results indicate that large reductions in dopamine concentrations in the rat brain can produce modest but significant changes in the binding of radioligands to VMAT2, which can be reversed by replenishment of dopamine using exogenous L-DOPA.


Assuntos
Dopamina/metabolismo , Neostriado/metabolismo , Tetrabenazina/análogos & derivados , Animais , Radioisótopos de Carbono/química , Dopamina/deficiência , Masculino , Neostriado/citologia , Neostriado/efeitos dos fármacos , Ligação Proteica , Ratos , Tetrabenazina/química , Tetrabenazina/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , alfa-Metiltirosina/farmacologia
15.
Nucl Med Biol ; 34(5): 479-82, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17591547

RESUMO

The in vivo binding of N-[18F]fluoroethyl-piperidinyl benzilate ([18F]FEPB) to the muscarinic cholinergic receptor was measured in awake and anesthetized rats. Studies were done using an equilibrium infusion technique to provide estimates of specific binding as distribution volume ratios. Anesthesia with either isoflurane or sodium pentobarbital produced a significant (65-90%) increase of radiotracer binding in receptor-rich brain regions (striatum, cortex, hippocampus) relative to awake controls. Pretreatment of anesthetized animals with the acetylcholinesterase inhibitor phenserine produced no further increases in radioligand binding, in contrast to the large (>70%) increases previously observed in awake animals following drug treatment. These studies demonstrate that anesthesia can produce significant changes in baseline biochemical measures that can obscure even very large effects of pharmacological challenges.


Assuntos
Benzilatos/farmacocinética , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Isoflurano/administração & dosagem , Pentobarbital/administração & dosagem , Piperidinas/farmacocinética , Anestésicos Inalatórios/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Radioisótopos de Flúor/farmacocinética , Hipnóticos e Sedativos/administração & dosagem , Masculino , Taxa de Depuração Metabólica/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Distribuição Tecidual/efeitos dos fármacos
16.
Bioorg Med Chem ; 13(3): 869-75, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15653352

RESUMO

Four 18F-labeled acetylcholinesterase (AChE) substrates, (S)-N-[18F]fluoroethyl-2-piperidinemethyl acetate (1), (R)-N-[18F]fluoroethyl-3-pyrrolidinyl acetate (2), N-[18F]fluoroethyl-4-piperidinyl acetate (3), and (R)-N-[18F]fluoroethyl-3-piperidinyl acetate (4), were evaluated for in vivo blood and brain metabolism in mice, brain pharmacokinetics in rats monkeys (M. nemistrina) using PET imaging. All 18F-labeled compounds were compared to N-[11C]methyl-4-piperidinyl propionate (PMP). Compound 1 was completely metabolized within 1 min in mouse blood and brain. This compound had relatively fast regional brain pharmacokinetics and poor discrimination between brain regions with different AChE concentration. Compound 4 showed relatively slower blood metabolism and slower pharmacokinetics than compound 1 but again poor discrimination between brain regions. Both compounds 1 and 4 showed different kinetic profiles than PMP in PET studies. Compound 3 had the slowest blood metabolism and slower pharmacokinetics than PMP. Compound 2 showed highly encouraging characteristics with an in vivo metabolism rate, primate brain uptake, and regional brain pharmacokinetics similar to [11C]PMP. The apparent hydrolysis rate constant k3 in primate cortex was very close to that of [11C]PMP. This compound has potential to be a good PET radiotracer for measuring brain AChE activity. The longer lifetime of 18F would permit longer imaging times and allows preparation of radiotracer batches for multiple patients and delivery of the tracer to other facilities, making the technique more widely available to clinical investigators.


Assuntos
Acetilcolinesterase/metabolismo , Flúor/química , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacologia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Feminino , Haplorrinos , Hidrólise , Masculino , Camundongos , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato , Distribuição Tecidual
17.
Nucl Med Biol ; 30(5): 491-500, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12831986

RESUMO

A series of N-fluoroethylpiperidinyl (1), N-fluoroethylpiperidinemethyl (2) and N-fluoroethylpyrrolidinyl (3) esters were synthesized and examined as new (18)F-labeled radiotracers for measuring brain cholinesterase activity. The fluoroethyl group, instead of methyl group, results in slower in vitro enzymatic cleavage rates and higher selectivity for AChE. Based on metabolism in mouse blood and PET time-activity curves in rats, two radiotracers were identified as potential candidates for further in vivo evaluation in higher species.


Assuntos
Acetilcolinesterase/metabolismo , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Ésteres/química , Ésteres/farmacocinética , Piperidinas/química , Piperidinas/farmacocinética , Pirrolidinas/química , Pirrolidinas/farmacocinética , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Córtex Cerebral/diagnóstico por imagem , Corpo Estriado/diagnóstico por imagem , Electrophorus , Ésteres/sangue , Feminino , Cavalos , Hidrólise , Marcação por Isótopo/métodos , Taxa de Depuração Metabólica , Camundongos , Piperidinas/sangue , Ligação Proteica , Pirrolidinas/sangue , Cintilografia , Compostos Radiofarmacêuticos/sangue , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
18.
Nucl Med Biol ; 30(3): 293-302, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12745021

RESUMO

The N-[(11)C]methylpiperidinyl esters are used as radiopharmaceuticals for measuring brain cholinesterase activity. We have synthesized a series of N-methylpiperidinemethyl (1), N-methylpyrrolidinyl (2) and N-methylpyrrolidinemethyl (3) esters and examined the effects of sterric constraint and stereochemistry on cholinesterase-mediated cleavage. Acetylcholinesterase exhibited a preference for primary esters 1 and for the R-isomers of both 1 and 2. Biological data for (S)-N-[(11)C]methyl-2-piperidinemethyl acetate (1a) were similar to [(11)C]AMP. These data better define the structure-activity relationships for cholinesterase radiotracers and provide lead compounds for (18)F- labeling.


Assuntos
Acetilcolinesterase/metabolismo , Encéfalo/efeitos dos fármacos , Piperidinas/síntese química , Pirrolidinas/síntese química , Compostos Radiofarmacêuticos/síntese química , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Ésteres , Feminino , Hidrólise , Camundongos , Piperidinas/metabolismo , Piperidinas/farmacologia , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacologia , Estereoisomerismo , Relação Estrutura-Atividade , Tomografia Computadorizada de Emissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA