Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 193: 106438, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38365045

RESUMO

Huntington's disease (HD) is a progressive neurodegenerative disease affecting motor and cognitive abilities. Multiple studies have found white matter anomalies in HD-affected humans and animal models of HD. The identification of sensitive white-matter-based biomarkers in HD animal models will be important in understanding disease mechanisms and testing the efficacy of therapeutic interventions. Here we investigated the progression of white matter deficits in the knock-in zQ175DN heterozygous (HET) mouse model of HD at 3, 6 and 11 months of age (M), reflecting different states of phenotypic progression. We compared findings from traditional diffusion tensor imaging (DTI) and advanced fixel-based analysis (FBA) diffusion metrics for their sensitivity in detecting white matter anomalies in the striatum, motor cortex, and segments of the corpus callosum. FBA metrics revealed progressive and widespread reductions of fiber cross-section and fiber density in myelinated bundles of HET mice. The corpus callosum genu was the most affected structure in HET mice at 6 and 11 M based on the DTI and FBA metrics, while the striatum showed the earliest progressive differences starting at 3 M based on the FBA metrics. Overall, FBA metrics detected earlier and more prominent alterations in myelinated fiber bundles compared to the DTI metrics. Luxol fast blue staining showed no loss in myelin density, indicating that diffusion anomalies could not be explained by myelin reduction but diffusion anomalies in HET mice were accompanied by increased levels of neurofilament light chain protein at 11 M. Altogether, our findings reveal progressive alterations in myelinated fiber bundles that can be measured using diffusion MRI, representing a candidate noninvasive imaging biomarker to study phenotype progression and the efficacy of therapeutic interventions in zQ175DN mice. Moreover, our study exposed higher sensitivity of FBA than DTI metrics, suggesting a potential benefit of adopting these advanced metrics in other contexts, including biomarker development in humans.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Substância Branca , Humanos , Animais , Camundongos , Imagem de Tensor de Difusão , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/genética , Imagem de Difusão por Ressonância Magnética , Substância Branca/diagnóstico por imagem , Modelos Animais de Doenças , Biomarcadores
2.
J Neurosci ; 43(49): 8275-8293, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-38073598

RESUMO

Neurodevelopmental disorders (NDDs) are a group of complex neurologic and psychiatric disorders. Functional and molecular imaging techniques, such as resting-state functional magnetic resonance imaging (rs-fMRI) and positron emission tomography (PET), can be used to measure network activity noninvasively and longitudinally during maturation in both humans and rodent models. Here, we review the current knowledge on rs-fMRI and PET biomarkers in the study of normal and abnormal neurodevelopment, including intellectual disability (ID; with/without epilepsy), autism spectrum disorder (ASD), and attention deficit hyperactivity disorder (ADHD), in humans and rodent models from birth until adulthood, and evaluate the cross-species translational value of the imaging biomarkers. To date, only a few isolated studies have used rs-fMRI or PET to study (abnormal) neurodevelopment in rodents during infancy, the critical period of neurodevelopment. Further work to explore the feasibility of performing functional imaging studies in infant rodent models is essential, as rs-fMRI and PET imaging in transgenic rodent models of NDDs are powerful techniques for studying disease pathogenesis, developing noninvasive preclinical imaging biomarkers of neurodevelopmental dysfunction, and evaluating treatment-response in disease-specific models.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Transtorno do Espectro Autista , Epilepsia , Lactente , Humanos , Adulto , Transtorno do Espectro Autista/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Tomografia por Emissão de Pósitrons , Biomarcadores , Encéfalo/diagnóstico por imagem
3.
EJNMMI Phys ; 10(1): 78, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38052966

RESUMO

BACKGROUND: Kinetic modeling in positron emission tomography (PET) requires measurement of the tracer plasma activity in the absence of a suitable reference region. To avoid invasive blood sampling, the use of an image derived input function has been proposed. However, an accurate delineation of the blood pool region in the PET image is necessary to obtain unbiased blood activity. Here, to perform brain kinetic modeling in [18F]SynVesT-1 dynamic scans, we make use of non-negative matrix factorization (NMF) to unmix the activity signal from the different tissues that can contribute to the heart region activity, and extract only the left ventricle activity in an unbiased way. This method was implemented in dynamic [18F]SynVesT-1 scans of mice anesthetized with either isoflurane or ketamine-xylazine, two anesthestics that we showed to affect differently radiotracer kinetics. The left ventricle activity (NMF-IDIF) and a manually delineated cardiac activity (IDIF) were compared with arterial blood samples (ABS), and for isoflurane anesthetized mice, arteriovenous (AV) shunt blood data were compared as well. Finally, brain regional 2 tissue compartment modeling was performed using IDIF and NMF-IDIF, and the model fit accuracy (weighted symmetrical mean absolute percentage error, wsMAPE) as well as the total volume of distribution (VT) were compared. RESULTS: In isoflurane anesthetized mice, the difference between ABS and NMF-IDIF activity (+ 12.8 [Formula: see text] 11%, p = 0.0023) was smaller than with IDIF (+ 16.4 [Formula: see text] 9.8%, p = 0.0008). For ketamine-xylazine anesthetized mice the reduction in difference was larger (NMF-IDIF: 16.9 [Formula: see text] 10%, p = 0.0057, IDIF: 56.3 [Formula: see text] 14%, p < 0.0001). Correlation coefficient between isoflurane AV-shunt time activity curves and NMF-IDIF (0.97 [Formula: see text] 0.01) was higher than with IDIF (0.94 [Formula: see text] 0.03). The brain regional 2TCM wsMAPE was improved using NMF-IDIF compared with IDIF, in isoflurane (NMF-IDIF: 1.24 [Formula: see text] 0.24%, IDIF: 1.56 [Formula: see text] 0.30%) and ketamine-xylazine (NMF-IDIF: 1.40 [Formula: see text] 0.24, IDIF: 2.62 [Formula: see text] 0.27) anesthetized mice. Finally, brain VT was significantly (p < 0.0001) higher using NMF-IDIF compared with IDIF, in isoflurane (3.97 [Formula: see text] 0.13% higher) and ketamine-xylazine (32.7 [Formula: see text] 2.4% higher) anesthetized mice. CONCLUSIONS: Image derived left ventricle blood activity calculated with NMF improves absolute activity quantification, and reduces the error in the kinetic modeling fit. These improvements are more pronounced in ketamine-xylazine than in isoflurane anesthetized mice.

5.
J Nucl Med ; 64(10): 1581-1587, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37591545

RESUMO

Huntington disease (HD) is a neurodegenerative disorder caused by an expanded polyglutamine (CAG) trinucleotide expansion in the huntingtin (HTT) gene that encodes the mutant huntingtin protein (mHTT). Visualization and quantification of cerebral mHTT will provide a proxy for target engagement and a means to evaluate therapeutic interventions aimed at lowering mHTT in the brain. Here, we validated the novel radioligand 11C-labeled 6-(5-((5-methoxypyridin-2-yl)methoxy)benzo[d]oxazol-2-yl)-2-methylpyridazin-3(2H)-one (11C-CHDI-180R) using PET imaging to quantify cerebral mHTT aggregates in a macaque model of HD. Methods: Rhesus macaques received MRI-guided intrastriatal delivery of a mixture of AAV2 and AAV2.retro viral vectors expressing an HTT fragment bearing 85 CAG repeats (85Q, n = 5), a control HTT fragment bearing 10 CAG repeats (10Q, n = 4), or vector diluent only (phosphate-buffered saline, n = 5). Thirty months after surgery, 90-min dynamic PET/CT imaging was used to investigate 11C-CHDI-180R brain kinetics, along with serial blood sampling to measure input function and stability of the radioligand. The total volume of distribution was calculated using a 2-tissue-compartment model as well as Logan graphical analysis for regional quantification. Immunostaining for mHTT was performed to corroborate the in vivo findings. Results: 11C-CHDI-180R displayed good metabolic stability (51.4% ± 4.0% parent in plasma at 60 min after injection). Regional time-activity curves displayed rapid uptake and reversible binding, which were described by a 2-tissue-compartment model. Logan graphical analysis was associated with the 2-tissue-compartment model (r 2 = 0.96, P < 0.0001) and used to generate parametric volume of distribution maps. Compared with controls, animals administered the 85Q fragment exhibited significantly increased 11C-CHDI-180R binding in several cortical and subcortical brain regions (group effect, P < 0.0001). No difference in 11C-CHDI-180R binding was observed between buffer and 10Q animals. The presence of mHTT aggregates in the 85Q animals was confirmed histologically. Conclusion: We validated 11C-CHDI-180R as a radioligand to visualize and quantify mHTT aggregated species in a HD macaque model. These findings corroborate our previous work in rodent HD models and show that 11C-CHDI-180R is a promising tool to assess the mHTT aggregate load and the efficacy of therapeutic strategies.


Assuntos
Doença de Huntington , Animais , Doença de Huntington/metabolismo , Proteína Huntingtina/genética , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Macaca mulatta/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tomografia por Emissão de Pósitrons , Modelos Animais de Doenças
6.
J Cereb Blood Flow Metab ; 43(9): 1612-1624, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37113068

RESUMO

We investigated the effect of isoflurane and ketamine-xylazine anesthesia on the positron emission tomography (PET) tracer [18F]SynVesT-1 in the mouse brain. [18F]SynVesT-1 PET scans were performed in C57BL/6J mice in five conditions: isoflurane anesthesia (ANISO), ketamine-xylazine (ANKX), awake freely moving (AW), awake followed by isoflurane administration (AW/ANISO) or followed by ketamine-xylazine (AW/ANKX) 20 min post tracer injection. ANISO, ANKX and AW scans were also performed in mice administered with levetiracetam (LEV, 200 mg/kg) to assess non-displaceable binding. Metabolite analysis was performed in ANISO, ANKX and AW mice. Finally, in vivo autoradiography in ANISO, ANKX and AW mice at 30 min post-injection was performed for validation. Kinetic modeling, with a metabolite corrected image derived input function, was performed to calculate total and non-displaceable volume of distribution (VT(IDIF)). VT(IDIF) was higher in ANISO compared to AW (p < 0.0001) while VT(IDIF) in ANKX was lower compared with AW (p < 0.0001). Non-displaceable VT(IDIF) was significantly different between ANISO and AW, but not between ANKX and AW. Change in the TAC washout was observed after administration of either isoflurane or ketamine-xylazine. Observed changes in tracer kinetics and volume of distribution might be explained by physiological changes due to anesthesia, as well as by induced cellular effects.


Assuntos
Isoflurano , Ketamina , Animais , Camundongos , Ketamina/farmacologia , Ketamina/metabolismo , Isoflurano/farmacologia , Xilazina/farmacologia , Xilazina/metabolismo , Camundongos Endogâmicos C57BL , Encéfalo/metabolismo
7.
Mov Disord ; 38(1): 143-147, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36544385

RESUMO

BACKGROUND: Dopamine system dysfunction and altered glucose metabolism are implicated in Huntington's disease (HD), a neurological disease caused by mutant huntingtin (mHTT) expression. OBJECTIVE: The aim was to characterize alterations in cerebral dopamine D2 /D3 receptor density and glucose utilization in a newly developed AAV-mediated NHP model of HD that expresses mHTT throughout numerous brain regions. METHODS: Positron emission tomography (PET) imaging was performed using [18 F]fallypride to quantify D2 /D3 receptor density and 2-[18 F]fluoro-2-deoxy-d-glucose ([18 F]FDG) to measure cerebral glucose utilization in these HD macaques. RESULTS: Compared to controls, HD macaques showed significantly reduced dopamine D2 /D3 receptor densities in basal ganglia (P < 0.05). In addition, HD macaques displayed significant glucose hypometabolism throughout the cortico-basal ganglia network (P < 0.05). CONCLUSIONS: [18 F]Fallypride and [18 F]FDG are PET imaging biomarkers of mHTT-mediated disease progression that can be used as noninvasive outcome measures in future therapeutic studies with this AAV-mediated HD macaque model. © 2022 International Parkinson and Movement Disorder Society.


Assuntos
Fluordesoxiglucose F18 , Doença de Huntington , Animais , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/metabolismo , Receptores de Dopamina D3/metabolismo , Dopamina/metabolismo , Macaca/metabolismo , Tomografia por Emissão de Pósitrons , Glucose/metabolismo
8.
J Med Chem ; 66(1): 641-656, 2023 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-36548390

RESUMO

Therapeutic interventions are being developed for Huntington's disease (HD), a hallmark of which is mutant huntingtin protein (mHTT) aggregates. Following the advancement to human testing of two [11C]-PET ligands for aggregated mHTT, attributes for further optimization were identified. We replaced the pyridazinone ring of CHDI-180 with a pyrimidine ring and minimized off-target binding using brain homogenate derived from Alzheimer's disease patients. The major in vivo metabolic pathway via aldehyde oxidase was blocked with a 2-methyl group on the pyrimidine ring. A strategically placed ring-nitrogen on the benzoxazole core ensured high free fraction in the brain without introducing efflux. Replacing a methoxy pendant with a fluoro-ethoxy group and introducing deuterium atoms suppressed oxidative defluorination and accumulation of [18F]-signal in bones. The resulting PET ligand, CHDI-650, shows a rapid brain uptake and washout profile in non-human primates and is now being advanced to human testing.


Assuntos
Doença de Huntington , Tomografia por Emissão de Pósitrons , Animais , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Ligantes , Tomografia por Emissão de Pósitrons/métodos , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/tratamento farmacológico , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo
9.
Neuroimage ; 264: 119771, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36436710

RESUMO

BACKGROUND: Synaptic vesicle glycoprotein 2A (SV2A) is a vesicle glycoprotein involved in neurotransmitter release. SV2A is located on the pre-synaptic terminals of neurons and visualized using the radioligand [11C]UCB-J and positron emission tomography (PET) imaging. Thus, SV2A PET imaging can provide a proxy for pre-synaptic density in health and disease. This study aims to apply independent component analysis (ICA) to SV2A PET data acquired in mice to identify pre-synaptic density networks (pSDNs), explore how ageing affects these pSDNs, and determine the impact of a neurological disorder on these networks. METHODS: We used [11C]UCB-J PET imaging data (n = 135) available at different ages (3, 7, 10, and 16 months) in wild-type (WT) C57BL/6J mice and in diseased mice (mouse model of Huntington's disease, HD) with reported synaptic deficits. First, ICA was performed on a healthy dataset after it was split into two equal-sized samples (n = 36 each) and the analysis was repeated 50 times in different partitions. We tested different model orders (8, 12, and 16) and identified the pSDNs. Next, we investigated the effect of age on the loading weights of the identified pSDNs. Additionally, the identified pSDNs were compared to those of diseased mice to assess the impact of disease on each pSDNs. RESULTS: Model order 12 resulted in the preferred choice to provide six reliable and reproducible independent components (ICs) as supported by the cluster-quality index (IQ) and regression coefficients (ß) values. Temporal analysis showed age-related statistically significant changes on the loading weights in four ICs. ICA in an HD model revealed a statistically significant disease-related effect on the loading weights in several pSDNs in line with the progression of the disease. CONCLUSION: This study validated the use of ICA on SV2A PET data acquired with [11C]UCB-J for the identification of cerebral pre-synaptic density networks in mice in a rigorous and reproducible manner. Furthermore, we showed that different pSDNs change with age and are affected in a disease condition. These findings highlight the potential value of ICA in understanding pre-synaptic density networks in the mouse brain.


Assuntos
Glicoproteínas de Membrana , Pirrolidinonas , Animais , Camundongos , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Tomografia por Emissão de Pósitrons/métodos , Envelhecimento , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo
10.
Front Neurosci ; 16: 901091, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35645721

RESUMO

The linear parametric neurotransmitter positron emission tomography (lp-ntPET) kinetic model can be used to detect transient changes (activation) in endogenous neurotransmitter levels. Preclinical PET scans in awake animals can be performed to investigate neurotransmitter transient changes. Here we use the spatiotemporal kernel reconstruction (Kernel) for noise reduction in dynamic PET, and lp-ntPET kinetic modeling. Kernel is adapted for motion correction reconstruction, applied in awake rat PET scans. We performed 2D rat brain phantom simulation using the ntPET model at 3 different noise levels. Data was reconstructed with independent frame reconstruction (IFR), IFR with HYPR denoising, and Kernel, and lp-ntPET kinetic parameters (k 2a : efflux rate, γ: activation magnitude, t d : activation onset time, and t p : activation peak time) were calculated. Additionally, significant activation magnitude (γ) difference with respect to a region with no activation (rest) was calculated. Finally, [11C]raclopride experiments were performed in anesthetized and awake rats, injecting cold raclopride at 20 min after scan start to simulate endogenous neurotransmitter release. For simulated data at the regional level, IFR coefficient of variation (COV) of k 2a , γ, t d and t p was reduced with HYPR denoising, but Kernel showed the lowest COV (2 fold reduction compared with IFR). At the pixel level the same trend is observed for k 2a , γ, t d and t p COV, but reduction is larger with Kernel compared with IFR (10-14 fold). Bias in γ with respect with noise-free values was additionally reduced using Kernel (difference of 292, 72.4, and -6.92% for IFR, IFR+KYPR, and Kernel, respectively). Significant difference in activation between the rest and active region could be detected at a simulated activation of 160% for IFR and IFR+HYPR, and of 120% for Kernel. In rat experiments, lp-ntPET parameters have better confidence intervals using Kernel. In the γ, and t d parametric maps, the striatum structure can be identified with Kernel but not with IFR. Striatum voxel-wise γ, t d and t p values have lower variability using Kernel compared with IFR and IFR+HYPR. The spatiotemporal kernel reconstruction adapted for motion correction reconstruction allows to improve lp-ntPET kinetic modeling noise in awake rat studies, as well as detection of subtle neurotransmitter activations.

11.
J Cereb Blood Flow Metab ; 42(10): 1867-1878, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35570828

RESUMO

Alterations in synaptic vesicle glycoprotein 2 A (SV2A) have been associated with several neuropsychiatric and neurodegenerative disorders. Therefore, SV2A positron emission tomography (PET) imaging may provide a unique tool to investigate synaptic density dynamics during disease progression and after therapeutic intervention. This study aims to extensively characterize the novel radioligand [18F]SynVesT-1 for preclinical applications. In C57Bl/6J mice (n = 39), we assessed the plasma profile of [18F]SynVesT-1, validated the use of a noninvasive image-derived input function (IDIF) compared to an arterial input function (AIF), performed a blocking study with levetiracetam (50 and 200 mg/kg, i.p.) to verify the specificity towards SV2A, examined kinetic models for volume of distribution (VT) quantification, and explored test-retest reproducibility of [18F]SynVesT-1 in the central nervous system (CNS). Plasma availability of [18F]SynVesT-1 decreased rapidly (13.4 ± 1.5% at 30 min post-injection). VT based on AIF and IDIF showed excellent agreement (r2 = 0.95, p < 0.0001) and could be reliably estimated with a 60-min acquisition. The blocking study resulted in a complete blockade with no suitable reference region. Test-retest analysis indicated good reproducibility (mean absolute variability <10%). In conclusion, [18F]SynVesT-1 is selective for SV2A with optimal kinetics representing a candidate tool to quantify CNS synaptic density non-invasively.


Assuntos
Encéfalo , Vesículas Sinápticas , Animais , Encéfalo/metabolismo , Glicoproteínas/metabolismo , Levetiracetam , Camundongos , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/metabolismo , Reprodutibilidade dos Testes , Vesículas Sinápticas/metabolismo
12.
Sci Transl Med ; 14(630): eabm3682, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35108063

RESUMO

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by a CAG trinucleotide expansion in the huntingtin (HTT) gene that encodes the pathologic mutant HTT (mHTT) protein with an expanded polyglutamine (polyQ) tract. Whereas several therapeutic programs targeting mHTT expression have advanced to clinical evaluation, methods to visualize mHTT protein species in the living brain are lacking. Here, we demonstrate the development and characterization of a positron emission tomography (PET) imaging radioligand with high affinity and selectivity for mHTT aggregates. This small molecule radiolabeled with 11C ([11C]CHDI-180R) allowed noninvasive monitoring of mHTT pathology in the brain and could track region- and time-dependent suppression of mHTT in response to therapeutic interventions targeting mHTT expression in a rodent model. We further showed that in these animals, therapeutic agents that lowered mHTT in the striatum had a functional restorative effect that could be measured by preservation of striatal imaging markers, enabling a translational path to assess the functional effect of mHTT lowering.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Animais , Corpo Estriado/diagnóstico por imagem , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/genética , Doença de Huntington/metabolismo , Ligantes , Doenças Neurodegenerativas/patologia
13.
J Nucl Med ; 63(8): 1245-1251, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35027368

RESUMO

Traumatic spinal cord injury (SCI) is a neurologic condition characterized by long-term motor and sensory neurologic deficits as a consequence of an external physical impact damaging the spinal cord. Anatomic MRI is considered the gold-standard diagnostic tool to obtain structural information for the prognosis of acute SCI; however, it lacks functional objective information to assess SCI progression and recovery. In this study, we explored the use of synaptic vesicle glycoprotein 2A (SV2A) PET imaging to detect spinal cord lesions noninvasively after SCI. Methods: Mice (n = 7) and rats (n = 8) subjected to unilateral moderate cervical (C5) contusion were euthanized 1 wk after SCI for histologic and autoradiographic (3H-labeled (4R)-1-[(3-methylpyridin-4-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one [UCB-J]) investigation of SV2A levels. Longitudinal 11C-UCB-J PET/CT imaging was performed in sham (n = 7) and SCI rats (n = 8) 1 wk and 6 wk after SCI. Animals also underwent an 18F-FDG PET scan during the latter time point. Postmortem tissue SV2A analysis to corroborate in vivo PET findings was performed 6 wk after SCI. Results: A significant SV2A loss (ranging from -70.3% to -87.3%; P < 0.0001) was measured at the epicenter of the impact in vitro in both mouse and rat contusion SCI models. Longitudinal 11C-UCB-J PET imaging detected SV2A loss in SCI rats (-49.0% ± 8.1% at 1 wk and -52.0% ± 12.9% at 6 wk after SCI), with no change observed in sham rats. In contrast, 18F-FDG PET imaging measured only subtle hypometabolism (-17.6% ± 14.7%). Finally, postmortem 3H-UCB-J autoradiography correlated with the in vivo SV2A PET findings (r = 0.92, P < 0.0001). Conclusion:11C-UCB-J PET/CT imaging is a noninvasive marker for SV2A loss after SCI. Collectively, these findings indicate that SV2A PET may provide an objective measure of SCI and thus represent a valuable tool to evaluate novel therapeutics. Clinical assessment of SCI with SV2A PET imaging is highly recommended.


Assuntos
Contusões , Traumatismos da Medula Espinal , Animais , Biomarcadores , Fluordesoxiglucose F18 , Glicoproteínas de Membrana , Camundongos , Modelos Teóricos , Proteínas do Tecido Nervoso , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia por Emissão de Pósitrons/métodos , Pirrolidinonas/química , Ratos , Traumatismos da Medula Espinal/diagnóstico por imagem
14.
J Nucl Med ; 63(6): 942-947, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34531262

RESUMO

Synaptic dysfunction is a primary mechanism underlying Huntington disease (HD) progression. This study investigated changes in synaptic vesicle glycoprotein 2A (SV2A) density by means of 11C-UCB-J small-animal PET imaging in the central nervous system of mice with HD. Methods: Dynamic 11C-UCB-J small-animal PET imaging was performed at clinically relevant disease stages (at 3, 7, 10, and 16 mo) in the heterozygous knock-in Q175DN mouse model of HD and wild-type littermates (16-18 mice per genotype and time point). Cerebral 11C-UCB-J analyses were performed to assess genotypic differences during presymptomatic (3 mo) and symptomatic (7-16 mo) disease stages. 11C-UCB-J binding in the spinal cord was quantified at 16 mo. 3H-UCB-J autoradiography and SV2A immunofluorescence were performed postmortem in mouse and human brain tissues. Results:11C-UCB-J binding was lower in symptomatic heterozygous mice than in wild-type littermates in parallel with disease progression (7 and 10 mo: P < 0.01; 16 mo: P < 0.0001). Specific 11C-UCB-J binding was detectable in the spinal cord, with symptomatic heterozygous mice displaying a significant reduction (P < 0.0001). 3H-UCB-J autoradiography and SV2A immunofluorescence corroborated the in vivo measurements demonstrating lower SV2A in heterozygous mice (P < 0.05). Finally, preliminary analysis of SV2A in the human brain postmortem suggested lower SV2A in HD gene carriers than in controls without dementia. Conclusion:11C-UCB-J PET detected SV2A deficits during symptomatic disease in heterozygous mice in both the brain and the spinal cord and therefore may be suitable as a novel marker of synaptic integrity widely distributed in the central nervous system. On clinical application, 11C-UCB-J PET imaging may have promise for SV2A measurement in patients with HD during disease progression and after disease-modifying therapeutic strategies.


Assuntos
Doença de Huntington , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Progressão da Doença , Humanos , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Piridinas/metabolismo , Vesículas Sinápticas/metabolismo
15.
Eur J Nucl Med Mol Imaging ; 49(4): 1166-1175, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34651228

RESUMO

PURPOSE: As several therapies aimed at lowering mutant huntingtin (mHTT) brain levels in Huntington's disease (HD) are currently being investigated, noninvasive positron emission tomography (PET) imaging of mHTT could be utilized to directly evaluate therapeutic efficacy and monitor disease progression. Here we characterized and longitudinally assessed the novel radioligand [11C]CHDI-626 for mHTT PET imaging in the zQ175DN mouse model of HD. METHODS: After evaluating radiometabolites and radioligand kinetics, we conducted longitudinal dynamic PET imaging at 3, 6, 9, and 13 months of age (M) in wild-type (WT, n = 17) and heterozygous (HET, n = 23) zQ175DN mice. Statistical analysis was performed to evaluate temporal and genotypic differences. Cross-sectional cohorts at each longitudinal time point were included for post-mortem [3H]CHDI-626 autoradiography. RESULTS: Despite fast metabolism and kinetics, the radioligand was suitable for PET imaging of mHTT. Longitudinal quantification could discriminate between genotypes already at premanifest stage (3 M), showing an age-associated increase in signal in HET mice in parallel with mHTT aggregate load progression, as supported by the post-mortem [3H]CHDI-626 autoradiography. CONCLUSION: With clinical evaluation underway, [11C]CHDI-626 PET imaging appears to be a suitable preclinical candidate marker to monitor natural HD progression and for the evaluation of mHTT-lowering therapies.


Assuntos
Doença de Huntington , Animais , Radioisótopos de Carbono , Estudos Transversais , Modelos Animais de Doenças , Humanos , Doença de Huntington/metabolismo , Camundongos , Tomografia por Emissão de Pósitrons/métodos
16.
Front Med (Lausanne) ; 8: 753977, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34746189

RESUMO

Preclinical brain positron emission tomography (PET) in animals is performed using anesthesia to avoid movement during the PET scan. In contrast, brain PET scans in humans are typically performed in the awake subject. Anesthesia is therefore one of the principal limitations in the translation of preclinical brain PET to the clinic. This review summarizes the available literature supporting the confounding effect of anesthesia on several PET tracers for neuroscience in preclinical small animal scans. In a second part, we present the state-of-the-art methodologies to circumvent this limitation to increase the translational significance of preclinical research, with an emphasis on motion correction methods. Several motion tracking systems compatible with preclinical scanners have been developed, each one with its advantages and limitations. These systems and the novel experimental setups they can bring to preclinical brain PET research are reviewed here. While technical advances have been made in this field, and practical implementations have been demonstrated, the technique should become more readily available to research centers to allow for a wider adoption of the motion correction technique for brain research.

17.
Neuroimage Clin ; 31: 102701, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34090124

RESUMO

Neuroinflammation is a key component of epileptogenesis, the process leading to acquired epilepsy. In recent years, with the development of non-invasive in vivo positron emission tomography (PET) imaging of translocator protein 18 kDa (TSPO), a marker of neuroinflammation, it has become possible to perform longitudinal studies to characterize neuroinflammation at different disease stages in animal models of epileptogenesis. This study aimed to utilize the prognostic capability of TSPO PET imaging at disease onset (2 weeks post-SE) to categorize epileptic rats with distinct seizure burden based on TSPO levels at disease onset and investigate their association to TSPO expression at the chronic epilepsy stage. Controls (n = 14) and kainic acid-induced status epilepticus (KASE) rats (n = 41) were scanned non-invasively with [18F]PBR111 PET imaging measuring TSPO expression. Animals were monitored using video-electroencephalography (vEEG) up to chronic disease (12 weeks post-SE), at which TSPO levels ([3H]PK11195) as well as other post-mortem abnormalities (namely synaptic density ([3H]UCB-J), neuronal loss (NeuN), and neurodegeneration (FjC)) were investigated. By applying multivariate analysis, TSPO PET imaging at disease onset identified three KASE groups with significantly different spontaneous recurrent seizures (SRS) burden (defined as rare SRS, sporadic SRS, and frequent SRS) (p = 0.003). Interestingly, TSPO levels were significantly different when comparing the three KASE groups (p < 0.0001), with the frequent SRS group characterized only by a limited focal TSPO increase at disease onset. On the contrary, TSPO measured during chronic epilepsy was found to be the highest in the frequent SRS group and correlated with seizure burden (r = 0.826, p < 0.0001). Importantly, early and chronic TSPO levels did not correlate (r = -0.05). Finally, significant pathological changes in neuronal loss, synaptic density, and neurodegeneration were found not only when compared to control animals (p < 0.01), but also between the three KASE rat categories in the hippocampus (p < 0.05). Early and chronic TSPO upregulation following epileptogenic insult appear to be driven by two superimposed dynamic processes. The former is associated with epileptogenesis as measured at disease onset, while the latter is related to seizure frequency as quantified during chronic epilepsy.


Assuntos
Epilepsia do Lobo Temporal , Receptores de GABA-A/metabolismo , Animais , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/diagnóstico por imagem , Epilepsia do Lobo Temporal/metabolismo , Imagem Molecular , Fenótipo , Tomografia por Emissão de Pósitrons , Ratos , Regulação para Cima
18.
Phys Med Biol ; 66(11)2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-33906176

RESUMO

Depending on the molar activity of the tracer, the maximal allowable injected activity in mouse brain PET studies can be extremely low in order to avoid receptor saturation. Therefore, a high level of noise can be present in the image. We investigate several dynamic PET reconstruction methods in reduced counts, or equivalently in reduced injected activity, data exemplified in [11C]racloprideBPNDandR1quantification using the simplified reference tissue model (SRTM). We compared independent frame reconstruction (IFR), post-reconstruction HYPR denoising (IFR + HYPR), direct reconstruction using the SRTM model (DIR-SRTM), and the spatial (KERS) and spatiotemporal kernel reconstruction (KERST). Additionally, HYPR denoising of the frames used as features for the calculation of the spatial kernel matrix, was investigated (KERS-HYPR and KERST-HYPR).In vivodata of 11 mice, was used to generate list-mode data for five reduced count levels corresponding to reductions by a factor 4, 8, 12, 16 and 32 (equivalently 2.07, 1.04, 0.691, 0.518, and 0.260 MBq). Correlation of regionalBPNDandR1values (reduced versus full counts reconstructions) was high (r > 0.94) for all methods, with KERS-HYPR and KERST-HYPR reaching the highest correlation (r > 0.96). Among methods with regularization, DIR-SRTM showed the largest variability inBPND(Bland-Altman SD from 3.0% to 12%), while IFR showed it forR1(5.1%-14.6%). KERST and KERST-HYPR were the only methods with Bland-Altman bias and SD below 5% for noise level up to a reduction factor of 16. At the voxel level,BPNDandR1correlation was gradually decreased with increasing noise, with the largest correlation (BPNDr > 0.88,R1r > 0.62) for KERS-HYPR and KERST-HYPR. The spatial and the spatiotemporal kernel methods performed similarly, while using only temporal regularization with direct reconstruction showed more variability. AlthoughR1 values present noise, using the spatiotemporal kernel reconstruction, accurate estimates of binding potential could be obtained with mouse injected activities as low as 0.26-0.518 MBq. This is desirable in order to maintain the tracer kinetics principle in mouse studies.


Assuntos
Processamento de Imagem Assistida por Computador , Tomografia por Emissão de Pósitrons , Algoritmos , Animais , Encéfalo/diagnóstico por imagem , Cinética , Camundongos , Racloprida
19.
Neuroimage ; 233: 117961, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33741482

RESUMO

Since accurate quantification of 2-deoxy-2-18F-fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) requires dynamic acquisition with arterial input function, more practical semi-quantitative (static) approaches are often preferred. However, static standardized uptake values (SUV) are typically biased due to large variations in body weight (BW) occurring over time in animal studies. This study aims to improve static [18F]FDG PET SUV quantification by better accounting for BW variations in rats. We performed dynamic [18F]FDG PET imaging with arterial blood sampling in rats (n = 27) with different BW (range 0.230-0.487 kg). By regressing the area under the curve of the input function divided by injected activity against BW (r2=0.697), we determined a conversion factor f(BW) to be multiplied with SUV and SUVglc to obtain ratSUV and ratSUVglc, providing an improved estimate of the net influx rate Ki (r = 0.758, p<0.0001) and cerebral metabolic rate of glucose MRglc (r = 0.906, p<0.0001), respectively. In conclusion, the proposed ratSUV and ratSUVglc provide a proxy for the Ki and MRglc based on a single static [18F]FDG PET SUV measurement improving clinical significance and translation of rodent studies. Given a defined strain, sex, age, diet, and weight range, this method is applicable for future experiments by converting SUV with the derived f(BW).


Assuntos
Encéfalo/metabolismo , Fluordesoxiglucose F18/metabolismo , Glucose/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Animais , Peso Corporal/fisiologia , Encéfalo/diagnóstico por imagem , Masculino , Ratos , Ratos Sprague-Dawley
20.
Mol Imaging Biol ; 23(2): 208-219, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33179158

RESUMO

PURPOSE: Our aim in this study was to compare different non-invasive pharmacokinetic models and assess test-retest reproducibility of the radioligand [11C]SCH23390 for the quantification of dopamine D1-like receptor (D1R) in both wild-type (WT) mice and heterozygous (HET) Q175DN mice as Huntington's disease (HD) model. PROCEDURES: Adult WT (n = 9) and HET (n = 14) mice underwent a 90-min [11C]SCH23390 positron emission tomography (PET) scan followed by computed tomography (CT) to evaluate the pharmacokinetic modelling in healthy and diseased conditions. Additionally, 5 WT mice and 7 HET animals received a second [11C]SCH23390 PET scan for test-retest reproducibility. Parallel assessment of the simplified reference tissue model (SRTM), the multilinear reference tissue model (MRTM) and the Logan reference tissue model (Logan Ref) using the striatum as a receptor-rich region and the cerebellum as a receptor-free (reference) region was performed to define the most suitable method for regional- and voxel-based quantification of the binding potential (BPND). Finally, standardised uptake value ratio (SUVR-1) was assessed as a potential simplified measurement. RESULTS: For all models, we measured a significant decline in dopamine D1R density (e.g. SRTM = - 38.5 ± 5.0 %, p < 0.0001) in HET mice compared to WT littermates. Shortening the 90-min scan duration resulted in large underestimation of striatal BPND in both WT mice (SRTM 60 min: - 17.7 ± 2.8 %, p = 0.0078) and diseased HET (SRTM 60 min: - 13.1 ± 4.1 %, p = 0.0001). Striatal BPND measurements were very reproducible with an average test-retest variability below 5 % when using both MRTM and SRTM. Parametric BPND maps generated with SRTM were highly reliable, showing nearly perfect agreement to the regional analysis (r2 = 0.99, p < 0.0001). Finally, SRTM provided the most accurate estimate for relative tracer delivery R1 with both regional- and voxel-based analyses. SUVR-1 at different time intervals were not sufficiently reliable when compared to BPND (r2 < 0.66). CONCLUSIONS: Ninety-minute acquisition and the use of SRTM for pharmacokinetic modelling is recommended. [11C]SCH23390 PET imaging demonstrates optimal characteristics for the study of dopamine D1R density in models of psychiatric and neurological disorders as exemplified in the Q175DN mouse model of HD.


Assuntos
Benzazepinas/farmacocinética , Encéfalo/diagnóstico por imagem , Doença de Huntington/diagnóstico por imagem , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Receptores de Dopamina D1/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Radioisótopos de Carbono , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Transgênicos , Receptores de Dopamina D1/metabolismo , Reprodutibilidade dos Testes , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...