Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Ther ; 25(11): 2513-2525, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28822689

RESUMO

A causal role of hypercholesterolemia in non-ischemic heart failure has never been demonstrated. Adeno-associated viral serotype 8 (AAV8)-low-density lipoprotein receptor (AAV8-LDLr) gene transfer was performed in LDLr-deficient mice without and with pressure overload induced by transverse aortic constriction (TAC). AAV8-LDLr gene therapy resulted in an 82.8% (p < 0.0001) reduction of plasma cholesterol compared with controls. Mortality rate was lower (p < 0.05) in AAV8-LDLr TAC mice compared with control TAC mice (hazard ratio for mortality 0.457, 95% confidence interval [CI] 0.237-0.882) during 8 weeks of follow-up. AAV8-LDLr gene therapy attenuated cardiac hypertrophy, reduced interstitial and perivascular fibrosis, and decreased lung congestion in TAC mice. Cardiac function, quantified by invasive hemodynamic measurements and magnetic resonance imaging, was significantly improved 8 weeks after sham operation or after TAC in AAV8-LDLr mice compared with respective control groups. Myocardial protein levels of mammalian target of rapamycin and of acetyl-coenzyme A carboxylase were strikingly decreased following cholesterol lowering in mice without and with pressure overload. AAV8-LDLr therapy potently reduced cardiac glucose uptake and counteracted metabolic remodeling following pressure overload. Furthermore, oxidative stress and myocardial apoptosis were decreased following AAV8-LDLr therapy in mice with pressure overload. In conclusion, cholesterol-lowering gene therapy potently counteracts structural and metabolic remodeling, and enhances cardiac function.


Assuntos
Cardiomegalia/terapia , Cardiomiopatias/terapia , Colesterol/metabolismo , Terapia Genética/métodos , Vetores Genéticos/metabolismo , Receptores de LDL/genética , Acetil-CoA C-Acetiltransferase/genética , Acetil-CoA C-Acetiltransferase/metabolismo , Animais , Aorta/cirurgia , Biomarcadores/metabolismo , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/mortalidade , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Cardiomiopatias/mortalidade , Constrição Patológica/complicações , Constrição Patológica/metabolismo , Constrição Patológica/patologia , Dependovirus/genética , Dependovirus/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Testes de Função Cardíaca , Hemodinâmica , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Receptores de LDL/deficiência , Análise de Sobrevida , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
2.
Int J Mol Sci ; 18(7)2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28718833

RESUMO

Studies evaluating the effects of high-saturated fat diets on cardiac function are most often confounded by diet-induced obesity and by systemic insulin resistance. We evaluated whether coconut oil, containing C12:0 and C14:0 as main fatty acids, aggravates pressure overload-induced cardiomyopathy induced by transverse aortic constriction (TAC) in C57BL/6 mice. Mortality rate after TAC was higher (p < 0.05) in 0.2% cholesterol 10% coconut oil diet-fed mice than in standard chow-fed mice (hazard ratio 2.32, 95% confidence interval 1.16 to 4.64) during eight weeks of follow-up. The effects of coconut oil on cardiac remodeling occurred in the absence of weight gain and of systemic insulin resistance. Wet lung weight was 1.76-fold (p < 0.01) higher in coconut oil mice than in standard chow mice. Myocardial capillary density (p < 0.001) was decreased, interstitial fibrosis was 1.88-fold (p < 0.001) higher, and systolic and diastolic function was worse in coconut oil mice than in standard chow mice. Myocardial glucose uptake was 1.86-fold (p < 0.001) higher in coconut oil mice and was accompanied by higher myocardial pyruvate dehydrogenase levels and higher acetyl-CoA carboxylase levels. The coconut oil diet increased oxidative stress. Myocardial triglycerides and free fatty acids were lower (p < 0.05) in coconut oil mice. In conclusion, coconut oil aggravates pressure overload-induced cardiomyopathy.


Assuntos
Cardiomiopatias/patologia , Óleo de Coco/efeitos adversos , Resistência à Insulina , Miocárdio/patologia , Obesidade/patologia , Pressão , Animais , Aorta/patologia , Peso Corporal , Capilares/patologia , Cardiomiopatias/sangue , Cardiomiopatias/complicações , Cardiomiopatias/fisiopatologia , Colesterol , Constrição Patológica , Diástole , Dieta , Feminino , Fibrose , Glucose/metabolismo , Hemodinâmica , Hipertrofia Ventricular Direita/complicações , Hipertrofia Ventricular Direita/patologia , Hipertrofia Ventricular Direita/fisiopatologia , Estimativa de Kaplan-Meier , Pulmão/patologia , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Transdução de Sinais , Sístole , Fator de Crescimento Transformador beta1/metabolismo
3.
Eur J Nucl Med Mol Imaging ; 43(12): 2219-2227, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27488857

RESUMO

PURPOSE: The cannabinoid type 2 receptor (CB2R) is expressed by immune cells such as monocytes and macrophages. In the brain, CB2R is primarily found on microglia. CB2R upregulation has been reported in animal models of Alzheimer's disease, with a preferential localization near amyloid beta (Aß) plaques, and in patients post mortem. We performed in vivo brain imaging and kinetic modelling of the CB2R tracer [11C]NE40 in healthy controls (HC) and in patients with Alzheimer's disease (AD) to investigate whether higher CB2R availability regionally colocalized to Aß deposits is present in vivo. METHODS: Dynamic 90-min [11C]NE40 PET scans were performed in eight HC and nine AD patients with full kinetic modelling using arterial sampling and metabolite correction and partial volume correction. All AD patients received a static [11C]PIB scan 40 min after injection. In four HC, a retest scan with [11C]NE40 PET was performed within 9 weeks to investigate test-retest characteristics. RESULTS: [11C]NE40 was metabolized quickly leading to 50 % of intact tracer 20 min after injection and 20 % at 90 min. A two-tissue kinetic model fitted most of the time-activity curves best; both binding potential (BPND) and distribution volume (V T) parameters could be used. Brain uptake was generally low with an average K 1 value of 0.07 ml/min/ml tissue. V T and BPND were in the range of 0.7 - 1.8 and 0.6 - 1.6, respectively. Test values in HC were about 30 % for V T and BPND. AD patients showed overall significantly lower CB2R binding. No relationship was found between regional or global amyloid load and CB2R availability. CONCLUSION: Kinetic modelling of [11C]NE40 is possible with a two-tissue reversible model. In contrast to preclinical and post-mortem data, [11C]NE40 PET shows lower CB2R availability in vivo in AD patients, with no relationship to Aß plaques. A possible explanation for these findings is that [11C]NE40 binds to CB2R with lower affinity and/or selectivity than to CB1R.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Modelos Neurológicos , Placa Amiloide/metabolismo , Quinolinas/farmacocinética , Receptor CB2 de Canabinoide/metabolismo , Disponibilidade Biológica , Simulação por Computador , Regulação para Baixo , Feminino , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Cinética , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
4.
Eur J Nucl Med Mol Imaging ; 43(1): 163-172, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26323575

RESUMO

PURPOSE: [(18)F]DPA-714 is a radiotracer with high affinity for TSPO. We have characterized the kinetics of [(18)F]DPA-714 in rat brain and evaluated its ability to quantify TSPO expression with PET using a neuroinflammation model induced by unilateral intracerebral injection of lipopolysaccharide (LPS). METHODS: Dynamic small-animal PET scans with [(18)F]DPA-714 were performed in Wistar rats on a FOCUS-220 system for up to 3 h. Both plasma and perfused brain homogenates were analysed using HPLC to quantify radiometabolites. Full kinetic modelling of [(18)F]DPA-714 brain uptake was performed using a metabolite-corrected arterial plasma input function. Binding potential (BPND) calculated as the distribution volume ratio minus one (DVR-1) between affected and healthy brain tissue was used as the outcome measure and evaluated against reference tissue models. RESULTS: The percentage of intact [(18)F]DPA-714 in arterial plasma samples was 92 ± 4 % at 10 min, 75 ± 8 % at 40 min and 52 ± 6 % at 180 min. The radiometabolite fraction in brain was negligible (<3 % at 30 min). Among the models investigated, the reversible two-tissue (2T) compartment model best described [(18)F]DPA-714 brain kinetics. BPND values obtained with a simplified and a multilinear reference tissue model (SRTM, MRTM) using the contralateral striatum as the reference region correlated well (Spearman's r = 0.96, p ≤ 0.003) with 2T BPND values calculated as DVR-1, and showed comparable bias (bias range 17.94 %, 20.32 %). Analysis of stability over time suggested that the acquisition time should be at least 90 min for SRTM and MRTM. CONCLUSION: Quantification of [(18)F]DPA-714 binding to TSPO with full kinetic modelling is feasible using a 2T model. SRTM and MRTM can be suggested as reasonable substitutes with the contralateral striatum as the reference region and a scan duration of at least 90 min. However, selection of the reference region depends on the disease model used.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Tomografia por Emissão de Pósitrons , Pirazóis , Pirimidinas , Receptores de GABA-A/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Radioisótopos de Flúor , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/diagnóstico por imagem , Inflamação/metabolismo , Injeções , Cinética , Ratos , Ratos Wistar
5.
Synapse ; 70(4): 153-62, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26799447

RESUMO

(18)F-FPEB is a promising PET tracer for studying the metabotropic glutamate subtype 5 receptor (mGluR5) expression in neuropsychiatric disorders. To assess the potential of (18)F-FPEB for longitudinal mGluR5 evaluation in patient studies, we evaluated the long-term test-retest reproducibility using various kinetic models in the human brain. Nine healthy volunteers underwent consecutive scans separated by a 6-month period. Dynamic PET was combined with arterial sampling and radiometabolite analysis. Total distribution volume (V(T)) and nondisplaceable binding potential (BP(ND)) were derived from a two-tissue compartment model without constraints (2TCM) and with constraining the K(1)/k(2) ratio to the value of either cerebellum (2TCM-CBL) or pons (2TCM-PONS). The effect of fitting different functions to the tracer parent fractions and reducing scan duration were assessed. Regional absolute test-retest variability (aTRV), coefficient of repeatability (CR) and intraclass correlation coefficient (ICC) were computed. The 2TCM-CBL showed best fits. The mean 6-month aTRV of V(T) ranged from 8 to 13% (CR < 25%) with ICC > 0.6 for all kinetic models. BPND from 2TCM-CBL with a sigmoid fit for the parent fractions showed the best reproducibility, with aTRV ≤ 7% (CR < 16%) and ICC > 0.9 in most regions. Reducing the scan duration from 90 to 60 min did not affect reproducibility. These results demonstrate for the first time that (18)F-FPEB brain PET has good long-term reproducibility, therefore validating its use to monitor mGluR5 expression in longitudinal clinical studies. We suggest a 2TCM-CBL with fitting a sigmoid function to the parent fractions to be optimal for this tracer.


Assuntos
Encéfalo/diagnóstico por imagem , Antagonistas de Aminoácidos Excitatórios/farmacocinética , Modelos Biológicos , Compostos Radiofarmacêuticos/farmacocinética , Receptor de Glutamato Metabotrópico 5/metabolismo , Adulto , Feminino , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons/normas , Ligação Proteica , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Reprodutibilidade dos Testes
6.
Stud Health Technol Inform ; 302: 972-976, 2023 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-37203547

RESUMO

Nowadays, the quantitative analysis of PET/CT data in patients with glioblastoma is not strictly standardized in the clinic and does not exclude the human factor. This study aimed to evaluate the relationship between the radiomic features of glioblastoma 11C-methionine PET images and the tumor-to-normal brain (T/N) ratio determined by radiologists in clinical routine. PET/CT data were obtained for 40 patients (mean age 55 ± 12 years; 77.5% men) with a histologically confirmed diagnosis of glioblastoma. Radiomic features were calculated for the whole brain and tumor-containing regions of interest using the RIA package for R. We redesigned the original RIA functions for GLCM and GLRLM calculation to reduce computation time significantly. Machine learning over radiomic features was applied to predict T/N with the best median correlation between the true and predicted values of 0.73 (p = 0.01). The present study showed a reproducible linear relationship between 11C-methionine PET radiomic features and a T/N indicator routinely assessed in brain tumors. Radiomics enabled utilizing texture properties of PET/CT neuroimaging that may reflect the biological activity of glioblastoma and can potentially augment the radiological assessment.


Assuntos
Glioblastoma , Masculino , Humanos , Adulto , Pessoa de Meia-Idade , Idoso , Feminino , Glioblastoma/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Radioisótopos de Carbono , Tomografia por Emissão de Pósitrons/métodos , Metionina , Estudos Retrospectivos
7.
Clin Nucl Med ; 47(8): 699-706, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35485864

RESUMO

OBJECTIVES: This study sought to assess 18 F-fludarabine ( 18 F-FLUDA) PET/CT's ability in differentiating primary central nervous system lymphomas (PCNSLs) from glioblastoma multiformes (GBMs). PATIENTS AND METHODS: Patients harboring either PCNSL (n = 8) before any treatment, PCNSL treated using corticosteroids (PCNSLh; n = 10), or GBM (n = 13) were investigated with conventional MRI and PET/CT, using 11 C-MET and 18 F-FLUDA. The main parameters measured with each tracer were SUV T and T/N ratios for the first 30 minutes of 11 C-MET acquisition, as well as at 3 different times after 18 F-FLUDA injection. The early 18 F-FLUDA uptake within the first minute of injection was equally considered, whereas this parameter was combined with the later uptakes to obtain R FLUDA 2 and R FLUDA 3 ratios. RESULTS: No significant differences in 11 C-MET uptakes were observed among PCNSL, PCNSLh, and GBM. With 18 F-FLUDA, a clear difference in dynamic GBM uptake was observed, which decreased over time after an early maximum, as compared with that of PCNSL, which steadily increased over time, PCNSLh exhibiting intermediate values. The most discriminative parameters consisting of R FLUDA 2 and R FLUDA 3 integrated the early tracer uptake (first 60 seconds), thereby provided 100% specificity and sensitivity. CONCLUSIONS: 18 F-FLUDA was shown to likely be a promising radiopharmaceutical for differentiating PCNSL from other malignancies, although a pretreatment with corticosteroids might compromise this differential diagnostic ability. The diagnostic role of 18 F-FLUDA should be further investigating, along with its potential of defining therapeutic strategies in patients with PCNSL, while assessing the treatments' effectiveness.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Linfoma , Corticosteroides , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Diagnóstico Diferencial , Fluordesoxiglucose F18 , Glioblastoma/diagnóstico por imagem , Glioblastoma/patologia , Humanos , Linfoma/diagnóstico por imagem , Linfoma/patologia , Metionina , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia por Emissão de Pósitrons , Vidarabina/análogos & derivados
8.
Aging (Albany NY) ; 11(17): 6872-6891, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31484164

RESUMO

Epidemiological studies support a strong association between non-high-density lipoprotein cholesterol levels and heart failure incidence. The objective of the current study was to evaluate the effect of selective cholesterol lowering adeno-associated viral serotype 8 (AAV8)-mediated low-density lipoprotein receptor (LDLr) gene transfer on cardiac remodelling and myocardial oxidative stress following transverse aortic constriction (TAC) in female C57BL/6 LDLr-/- mice with mild hypercholesterolemia. Cholesterol lowering gene transfer resulted in a 65.9% (p<0.0001) reduction of plasma cholesterol levels (51.2 ± 2.2 mg/dl) compared to controls (150 ± 7 mg/dl). Left ventricular wall area was 11.2% (p<0.05) lower in AAV8-LDLr TAC mice than in control TAC mice. In agreement, pro-hypertrophic myocardial proteins were potently decreased in AAV8-LDLr TAC mice. The degree of interstitial fibrosis and perivascular fibrosis was 31.0% (p<0.001) and 29.8% (p<0.001) lower, respectively, in AAV8-LDLr TAC mice compared to control TAC mice. These structural differences were associated with improved systolic and diastolic function and decreased lung congestion in AAV8-LDLr TAC mice compared to control TAC mice. Cholesterol lowering gene therapy counteracted myocardial oxidative stress and preserved the potential for myocardial fatty acid oxidation in TAC mice. In conclusion, cholesterol lowering gene therapy attenuates pressure overload-induced heart failure in mice with mild hypercholesterolemia.


Assuntos
Colesterol , Insuficiência Cardíaca , Hipercolesterolemia , Animais , Dependovirus , Terapia Genética , Vetores Genéticos , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/fisiopatologia , Hipercolesterolemia/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Estresse Oxidativo/fisiologia , Receptores de LDL/genética , Remodelação Ventricular/fisiologia
9.
Clin Transl Sci ; 11(4): 397-404, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29575526

RESUMO

Inhibition of fatty acid amide hydrolase (FAAH) potentiates endocannabinoid activity and is hypothesized to have therapeutic potential for mood and anxiety disorders and pain. The clinical profile of JNJ-42165279, an oral selective FAAH inhibitor, was assessed by investigating the pharmacokinetics, pharmacodynamics, safety, and binding to FAAH in the brain of healthy human volunteers. Concentrations of JNJ-42165279 (plasma, cerebrospinal fluid (CSF), urine) and fatty acid amides (FAA; plasma, CSF), and FAAH activity in leukocytes was determined in a phase I multiple ascending dose study. A positron emission tomography study with the FAAH tracer [11 C]MK3168 was conducted to determine brain FAAH occupancy after single and multiple doses of JNJ-42165279. JNJ-42165279 administration resulted in an increase in plasma and CSF FAA. Significant blocking of brain FAAH binding of [11 C]MK3168 was observed after pretreatment with JNJ-42165279. JNJ-42165279 produces potent central and peripheral FAAH inhibition. Saturation of brain FAAH occupancy occurred with doses ≥10 mg of JNJ-42165279. No safety concerns were identified.


Assuntos
Amidoidrolases/antagonistas & inibidores , Encéfalo/metabolismo , Endocanabinoides/metabolismo , Inibidores Enzimáticos/farmacologia , Piperazinas/farmacologia , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Amidoidrolases/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endocanabinoides/sangue , Endocanabinoides/líquido cefalorraquidiano , Feminino , Voluntários Saudáveis , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Traçadores Radioativos , Adulto Jovem
10.
EJNMMI Radiopharm Chem ; 1(1): 3, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29564380

RESUMO

BACKGROUND: Phosphodiesterase 10A (PDE10A) is an important regulator of nigrostriatal dopamine (DA) neurotransmission. However, little is known on the effect of alterations in DA neurotransmission on PDE10A availability. Here, we used [18F]JNJ42259152 PET to measure changes in PDE10A availability, secondary to pharmacological alterations in DA release and to investigate whether these are D1- or D2-receptor driven. RESULTS: Acute treatment of rats using D-amphetamine (5 mg, s.c. and 1 mg/kg i.v.) did not result in a significant change in PDE10A BPND compared to baseline conditions. 5-day D-amphetamine treatment (5 mg/kg, s.c.) increased striatal PDE10A BPND compared to the baseline (+24 %, p = 0.03). Treatment with the selective D2 antagonist SCH23390 (1 mg/kg) and D-amphetamine decreased PDE10A binding (-22 %, p = 0.03). Treatment with only SCH23390 further decreased PDE10A binding (-26 %, p = 0.03). No significant alterations in PDE10A mRNA levels were observed. CONCLUSIONS: Repeated D-amphetamine treatment significantly increased PDE10A binding, which is not observed upon selective D1 receptor blocking. This study suggests a potential pharmacological interaction between PDE10A enzymes and drugs modifying DA neurotransmission. Therefore, PDE10A binding in patients with neuropsychiatric disorders might be modulated by chronic DA-related treatment.

11.
J Cereb Blood Flow Metab ; 37(1): 97-105, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26661244

RESUMO

Studies in rodents suggest that flumazenil is a P-glycoprotein substrate at the blood-brain barrier. This study aimed to assess whether [11C]flumazenil is a P-glycoprotein substrate in humans and to what extent increased P-glycoprotein function in epilepsy may confound interpretation of clinical [11C]flumazenil studies used to assess gamma-aminobutyric acid A receptors. Nine drug-resistant patients with epilepsy and mesial temporal sclerosis were scanned twice using [11C]flumazenil before and after partial P-glycoprotein blockade with tariquidar. Volume of distribution, nondisplaceable binding potential, and the ratio of rate constants of [11C]flumazenil transport across the blood-brain barrier (K1/k2) were derived for whole brain and several regions. All parameters were compared between pre- and post-tariquidar scans. Regional results were compared between mesial temporal sclerosis and contralateral sides. Tariquidar significantly increased global K1/k2 (+23%) and volume of distribution (+10%), but not nondisplaceable binding potential. At the mesial temporal sclerosis side volume of distribution and nondisplaceable binding potential were lower in hippocampus (both ∼-19%) and amygdala (both ∼-16%), but K1/k2 did not differ, suggesting that only regional gamma-aminobutyric acid A receptor density is altered in epilepsy. In conclusion, although [11C]flumazenil appears to be a (weak) P-glycoprotein substrate in humans, this does not seem to affect its role as a tracer for assessing gamma-aminobutyric acid A receptor density.


Assuntos
Barreira Hematoencefálica/metabolismo , Epilepsia do Lobo Temporal/diagnóstico por imagem , Flumazenil/farmacocinética , Moduladores GABAérgicos/farmacocinética , Receptores de GABA-A/análise , Esclerose/diagnóstico por imagem , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adolescente , Adulto , Radioisótopos de Carbono , Resistência a Medicamentos , Humanos , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons/métodos , Tomografia por Emissão de Pósitrons/normas , Adulto Jovem
12.
J Nucl Med ; 57(9): 1436-41, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27199364

RESUMO

UNLABELLED: The P2X7 receptor (P2X7R) orchestrates neuroinflammation, and this is the basis for an increased interest in the development of antagonists inhibiting P2X7R function in the brain. This study provides the preclinical evaluation of (11)C-JNJ-54173717, a PET tracer for P2X7R in both rats and nonhuman primates. METHODS: (11)C-JNJ-54173717 is a high-affinity radiotracer for the human P2X7R (hP2X7R). Biodistribution and radiometabolite studies were performed. Viral vectors encoding either enhanced green fluorescent protein-hP2X7R or 3flag-hP2X7R were engineered and validated in cell culture. hP2X7R was regionally overexpressed in the rat striatum after stereotactic injection of viral vectors. Dynamic small-animal PET studies were performed in vector-injected rats and in healthy monkeys using (11)C-JNJ-54173717. RESULTS: The affinity of JNJ-54173717 was 1.6 ± 0.1 nM in a rat cortex P2X7R membrane binding assay. In a functional assay at the recombinant human and rat P2X7R orthologs, the half maximal inhibitory concentration (IC50) of JNJ-54173717 was 4.2 ± 0.01 nM and 7.6 ± 0.01 nM, respectively. The rat biodistribution study showed that (11)C-JNJ-54173717 crossed the blood-brain barrier and was cleared from plasma mainly via the hepatobiliary pathway. A polar radiometabolite was found in rat plasma. No radiometabolites were detected in rat brain. Dynamic small-animal PET showed binding of (11)C-JNJ-54173717 in the striatum expressing hP2X7R, with rapid washout from the noninjected control striatum and other brain regions. Likewise, (11)C-JNJ-54173717 PET signal was blocked by a chemically distinct P2X7R ligand, indicating specific binding to P2X7R in the monkey brain. CONCLUSION: JNJ-54173717 is a high-affinity P2X7R antagonist. An animal rat model stably expressing hP2X7R was developed and validated, identifying favorable characteristics for (11)C-JNJ-54173717 as a PET radioligand for in vivo visualization of hP2X7R. (11)C-JNJ-54173717 selectively visualized P2X7R in the monkey brain, and this radioligand will be further evaluated in a clinical setting to study P2X7R expression levels in neurodegenerative disorders.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Receptores Purinérgicos P2X7/metabolismo , Animais , Feminino , Humanos , Macaca mulatta , Masculino , Taxa de Depuração Metabólica , Especificidade de Órgãos , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Transgênicos , Ratos Wistar , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual , Regulação para Cima
13.
Nucl Med Biol ; 42(10): 753-61, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26220690

RESUMO

OBJECTIVE: The goal of this study was to measure functional and structural aspects of local neuroinflammation induced by intracerebral injection of lipopolysaccharide (LPS) in rats using TSPO microPET imaging with [(18)F]DPA-714, magnetic resonance imaging (MRI), in vitro autoradiography and immunohistochemistry (IHC) in order to characterize a small animal model for screening of new PET tracers targeting neuroinflammation. METHODS: Rats were injected stereotactically with LPS (50 µg) in the right striatum and with saline in the left striatum. [(18)F]DPA-714 microPET, MRI, in vitro autoradiography and IHC studies were performed at different time points after LPS injection for 1 month. RESULTS: Analysis of the microPET data demonstrated high uptake of the tracer in the LPS injected site with an affected-to-non-affected side-binding potential ratio (BPright-to-left) of 3.0 at 3 days after LPS injection. This BP ratio decreased gradually over time to 0.9 at 30 days after LPS injection. In vitro autoradiography ([(18)F]DPA-714) and IHC (CD68, GFAP and TSPO) confirmed local neuroinflammation in this model. Dynamic contrast enhanced (DCE) MRI demonstrated BBB breakdown near the LPS injection site at day 1, which gradually resolved over time and was absent at 1 month after LPS injection. CONCLUSION: The LPS model is useful for first screening of newly developed tracers because of the easy design and the robust, unilateral inflammatory reaction allowing the use of the contralateral region as control. Additionally, this model can be used to test and follow up the benefits of anti-inflammatory therapies by non-invasive imaging.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Proteínas de Transporte/metabolismo , Lipopolissacarídeos/farmacologia , Tomografia por Emissão de Pósitrons , Receptores de GABA-A/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Radioisótopos de Flúor , Inflamação/induzido quimicamente , Inflamação/diagnóstico por imagem , Inflamação/metabolismo , Injeções , Pirazóis , Pirimidinas , Ratos , Ratos Wistar
14.
J Nucl Med ; 56(10): 1520-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26229142

RESUMO

UNLABELLED: The quantification and variability of skeletal muscle glucose utilization (SMGU) in healthy subjects under basal (low insulin) conditions are poorly known. This information is essential early in clinical drug development to effectively interrogate novel pharmacologic interventions that modulate glucose uptake. The aim of this study was to determine test-retest characteristics and variability of SMGU within and between healthy subjects under basal conditions. Furthermore, different kinetic modeling strategies were evaluated to find the best-fitting model to assess SMGU studied by 18F-FDG. METHODS: Six healthy male volunteers underwent 2 dynamic 18F-FDG PET/CT scans with an interval of 24 h. Subjects were admitted to the clinical unit to minimize variability in daily activities and food intake and restrict physical activity. 18F-FDG PET/CT scans of gluteal and quadriceps muscle area were obtained with arterial input. Regions of interest were drawn over the muscle area to obtain time-activity curves and standardized uptake values (SUVs) between 60 and 90 min. Spectral analysis of the data and kinetic modeling was performed using 2-tissue-irreversible (2T3K), 2-tissue-reversible, and 3-tissue-sequential-irreversible (3T5KS) models. Reproducibility was assessed by intraclass correlation coefficients (ICCs) and within-subject coefficient of variation (WSCV). RESULTS: SUVs in gluteal and quadriceps areas were 0.56±0.09 and 0.64±0.07. ICCs (with 90% confidence intervals in parentheses) were 0.88 (0.64-0.96) and 0.96 (0.82-0.99), respectively, for gluteal and quadriceps muscles, and WSCV for gluteal and quadriceps muscles was 2.2% and 3.6%, respectively. The rate of glucose uptake into muscle was 0.0016±0.0004 mL/mL⋅min, with an ICC of 0.94 (0.93-0.95) and WSCV of 6.6% for the 3T5KS model, whereas an ICC of 0.98 (0.92-1.00) and WSCV of 2.8% was obtained for the 2T3K model. 3T5KS demonstrated the best fit to the measured experimental points. CONCLUSION: Minimal variability in skeletal muscle glucose uptake was observed under basal conditions in healthy subjects. SUV measurements and rate of glucose uptake values were reproducible, with an average WSCV of less than 5%. Compared with SUV, the 3-tissue model adds information about kinetics between blood, intra- and intercellular compartments, and phosphorylation that may highlight the exact mechanisms of metabolic changes after pharmacologic intervention.


Assuntos
Fluordesoxiglucose F18/farmacocinética , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Adulto , Feminino , Voluntários Saudáveis , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Compostos Radiofarmacêuticos , Reprodutibilidade dos Testes
15.
J Nucl Med ; 56(11): 1730-5, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26294297

RESUMO

UNLABELLED: Overexpression of the multidrug efflux transport P-glycoprotein may play an important role in pharmacoresistance. (11)C-laniquidar is a newly developed tracer of P-glycoprotein expression. The aim of this study was to develop a pharmacokinetic model for quantification of (11)C-laniquidar uptake and to assess its test-retest variability. METHODS: Two (test-retest) dynamic (11)C-laniquidar PET scans were obtained in 8 healthy subjects. Plasma input functions were obtained using online arterial blood sampling with metabolite corrections derived from manual samples. Coregistered T1 MR images were used for region-of-interest definition. Time-activity curves were analyzed using various plasma input compartmental models. RESULTS: (11)C-laniquidar was metabolized rapidly, with a parent plasma fraction of 50% at 10 min after tracer injection. In addition, the first-pass extraction of (11)C-laniquidar was low. (11)C-laniquidar time-activity curves were best fitted to an irreversible single-tissue compartment (1T1K) model using conventional models. Nevertheless, significantly better fits were obtained using 2 parallel single-tissue compartments, one for parent tracer and the other for labeled metabolites (dual-input model). Robust K1 results were also obtained by fitting the first 5 min of PET data to the 1T1K model, at least when 60-min plasma input data were used. For both models, the test-retest variability of (11)C-laniquidar rate constant for transfer from arterial plasma to tissue (K1) was approximately 19%. CONCLUSION: The accurate quantification of (11)C-laniquidar kinetics in the brain is hampered by its fast metabolism and the likelihood that labeled metabolites enter the brain. Best fits for the entire 60 min of data were obtained using a dual-input model, accounting for uptake of (11)C-laniquidar and its labeled metabolites. Alternatively, K1 could be obtained from a 5-min scan using a standard 1T1K model. In both cases, the test-retest variability of K1 was approximately 19%.


Assuntos
Benzazepinas/farmacocinética , Encéfalo/diagnóstico por imagem , Quinolinas/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Adulto , Biotransformação , Encéfalo/metabolismo , Radioisótopos de Carbono , Feminino , Voluntários Saudáveis , Humanos , Marcação por Isótopo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons/métodos , Reprodutibilidade dos Testes , Adulto Jovem
16.
Neurology ; 82(3): 279-81, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24353339

RESUMO

Phosphodiesterase 10A (PDE10A) belongs to a family of enzymes that hydrolyze cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate.(1) PDE10A is highly enriched in striatal medium spiny neurons (MSNs), where it regulates intracellular signaling.(1) PDE10A has been proposed as a therapeutic target for Huntington disease (HD), a disorder that preferentially affects MSNs, based on the observation that pharmacologic inhibition of PDE10A in transgenic HD mice significantly improved behavioral and neuropathologic abnormalities.(2) However, earlier work had shown that striatal PDE10A levels in HD mice already decline to minimal levels before onset of motor symptoms,(3) possibly because mutant huntingtin represses PDE10A transcription. Also, postmortem analysis of striatum of 3 patients with HD revealed strong reduction of PDE10A levels.(3) Depletion of PDE10A in HD striatum would at first sight seem hard to reconcile with a beneficial effect of PDE10A inhibitors in HD. However, a recent study reported a dramatic increase, rather than decrease, of PDE10A protein in MSNs of HD mice.(4) In light of these conflicting results and the strong interest in development of PDE10A inhibitors for clinical use in HD, it is important to determine whether PDE10A levels are affected in the striatum of patients with HD in vivo.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Animais , Corpo Estriado/patologia , Espinhas Dendríticas/patologia , Humanos , Doença de Huntington/patologia , Camundongos , Neurônios/patologia , Projetos Piloto , Pirazóis/administração & dosagem , Piridinas/administração & dosagem , Frações Subcelulares/patologia
17.
J Nucl Med ; 54(12): 2101-3, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24092938

RESUMO

UNLABELLED: Resistance to current drug therapy is an important issue in the treatment of epilepsy. Inadequate access of central nervous system drugs to their targets in the brain may be caused by overexpression or overactivity of multidrug transporters, such as P-glycoprotein (P-gp), at the blood-brain barrier. Laniquidar, an inhibitor of P-gp, has been labeled with (11)C for use in PET studies of P-gp expression in humans. Given potential interspecies differences in biodistribution, the purpose of this study was to ensure safe use of (11)C-laniquidar by determining the dosimetry of (11)C-laniquidar using whole-body PET studies. METHODS: Six healthy volunteers were subjected to a series of 10 whole-body PET scans within approximately 70 min. Five blood samples were taken during the series. RESULTS: High uptake of (11)C-laniquidar was seen in liver, spleen, kidneys, and lung, whereas brain uptake was low. The effective dose for (11)C-laniquidar was 4.76 ± 0.13 and 3.69 ± 0.01 µSv·MBq(-1) for women and men, respectively. CONCLUSION: Biodistribution and measured effective dose indicate that (11)C-laniquidar is a safe tracer for PET imaging, with a total dose of about 2 mSv for a brain PET/CT protocol.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Benzazepinas , Quinolinas , Doses de Radiação , Adulto , Benzazepinas/farmacocinética , Radioisótopos de Carbono , Feminino , Humanos , Masculino , Quinolinas/farmacocinética , Traçadores Radioativos , Radiometria
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA