Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Psychiatry Neurosci ; 49(3): E157-E171, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38692693

RESUMEN

BACKGROUND: Critical adolescent neural refinement is controlled by the DCC (deleted in colorectal cancer) protein, a receptor for the netrin-1 guidance cue. We sought to describe the effects of reduced DCC on neuroanatomy in the adolescent and adult mouse brain. METHODS: We examined neuronal connectivity, structural covariance, and molecular processes in a DCC-haploinsufficient mouse model, compared with wild-type mice, using new, custom analytical tools designed to leverage publicly available databases from the Allen Institute. RESULTS: We included 11 DCC-haploinsufficient mice and 16 wild-type littermates. Neuroanatomical effects of DCC haploinsufficiency were more severe in adolescence than adulthood and were largely restricted to the mesocorticolimbic dopamine system. The latter finding was consistent whether we identified the regions of the mesocorticolimbic dopamine system a priori or used connectivity data from the Allen Brain Atlas to determine de novo where these dopamine axons terminated. Covariance analyses found that DCC haploinsufficiency disrupted the coordinated development of the brain regions that make up the mesocorticolimbic dopamine system. Gene expression maps pointed to molecular processes involving the expression of DCC, UNC5C (encoding DCC's co-receptor), and NTN1 (encoding its ligand, netrin-1) as underlying our structural findings. LIMITATIONS: Our study involved a single sex (males) at only 2 ages. CONCLUSION: The neuroanatomical phenotype of DCC haploinsufficiency described in mice parallels that observed in DCC-haploinsufficient humans. It is critical to understand the DCC-haploinsufficient mouse as a clinically relevant model system.


Asunto(s)
Encéfalo , Receptor DCC , Dopamina , Haploinsuficiencia , Animales , Receptor DCC/genética , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/anatomía & histología , Dopamina/metabolismo , Ratones , Masculino , Expresión Génica , Vías Nerviosas , Factores de Edad , Femenino , Ratones Endogámicos C57BL , Envejecimiento/genética , Envejecimiento/fisiología
2.
BMC Neurol ; 21(1): 459, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34814867

RESUMEN

BACKGROUND: Dopamine transporter single-photon emission computed tomography (DaT-SPECT) can quantify the functional integrity of the dopaminergic nerve terminals and has been suggested as an imaging modality to verify the clinical diagnosis of Parkinson's disease (PD). Depending on the stage of progression, approximately 5-15% of participants clinically diagnosed with idiopathic PD have been observed in previous studies to have normal DaT-SPECT patterns. However, the utility of DaT-SPECT in enhancing early PD participant selection in a global, multicenter clinical trial of a potentially disease-modifying therapy is not well understood. METHODS: The SPARK clinical trial was a phase 2 trial of cinpanemab, a monoclonal antibody against alpha-synuclein, in participants with early PD. DaT-SPECT was performed at screening to select participants with DaT-SPECT patterns consistent with degenerative parkinsonism. Acquisition was harmonised across 82 sites. Images were reconstructed and qualitatively read at a central laboratory by blinded neuroradiologists for inclusion prior to automated quantitative analysis. RESULTS: In total, 482 unique participants were screened between January 2018 and May 2019; 3.8% (15/398) of imaged participants were excluded owing to negative DaT-SPECT findings (i.e., scans without evidence of dopaminergic deficit [SWEDD]). CONCLUSION: A smaller proportion of SPARK participants were excluded owing to SWEDD status upon DaT-SPECT screening than has been reported in prior studies. Further research is needed to understand the reasons for the low SWEDD rate in this study and whether these results are generalisable to future studies. If supported, the radiation risks, imaging costs, and operational burden of DaT-SPECT for enrichment may be mitigated by clinical assessment and other study design aspects. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03318523 . Date submitted: October 19, 2017. First Posted: October 24, 2017.


Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Enfermedad de Parkinson , Biomarcadores , Dopamina , Humanos , Enfermedad de Parkinson/diagnóstico por imagen , Tomografía Computarizada de Emisión de Fotón Único
3.
J Neurosci ; 38(20): 4655-4665, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29712788

RESUMEN

The axon guidance cue receptor DCC (deleted in colorectal cancer) plays a critical role in the organization of mesocorticolimbic pathways in rodents. To investigate whether this occurs in humans, we measured (1) anatomical connectivity between the substantia nigra/ventral tegmental area (SN/VTA) and forebrain targets, (2) striatal and cortical volumes, and (3) putatively associated traits and behaviors. To assess translatability, morphometric data were also collected in Dcc-haploinsufficient mice. The human volunteers were 20 DCC+/- mutation carriers, 16 DCC+/+ relatives, and 20 DCC+/+ unrelated healthy volunteers (UHVs; 28 females). The mice were 11 Dcc+/- and 16 wild-type C57BL/6J animals assessed during adolescence and adulthood. Compared with both control groups, the human DCC+/- carriers exhibited the following: (1) reduced anatomical connectivity from the SN/VTA to the ventral striatum [DCC+/+: p = 0.0005, r(effect size) = 0.60; UHV: p = 0.0029, r = 0.48] and ventral medial prefrontal cortex (DCC+/+: p = 0.0031, r = 0.53; UHV: p = 0.034, r = 0.35); (2) lower novelty-seeking scores (DCC+/+: p = 0.034, d = 0.82; UHV: p = 0.019, d = 0.84); and (3) reduced striatal volume (DCC+/+: p = 0.0009, d = 1.37; UHV: p = 0.0054, d = 0.93). Striatal volumetric reductions were also present in Dcc+/- mice, and these were seen during adolescence (p = 0.0058, d = 1.09) and adulthood (p = 0.003, d = 1.26). Together these findings provide the first evidence in humans that an axon guidance gene is involved in the formation of mesocorticolimbic circuitry and related behavioral traits, providing mechanisms through which DCC mutations might affect susceptibility to diverse neuropsychiatric disorders.SIGNIFICANCE STATEMENT Opportunities to study the effects of axon guidance molecules on human brain development have been rare. Here, the identification of a large four-generational family that carries a mutation to the axon guidance molecule receptor gene, DCC, enabled us to demonstrate effects on mesocorticolimbic anatomical connectivity, striatal volumes, and personality traits. Reductions in striatal volumes were replicated in DCC-haploinsufficient mice. Together, these processes might influence mesocorticolimbic function and susceptibility to diverse neuropsychiatric disorders.


Asunto(s)
Receptor DCC/genética , Sistema Límbico/fisiopatología , Vías Nerviosas/fisiopatología , Corteza Prefrontal/fisiopatología , Adulto , Envejecimiento/psicología , Animales , Axones , Conducta Exploratoria , Femenino , Heterocigoto , Humanos , Sistema Límbico/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Vías Nerviosas/diagnóstico por imagen , Trastornos de la Personalidad/genética , Trastornos de la Personalidad/psicología , Corteza Prefrontal/diagnóstico por imagen , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/psicología , Sustancia Negra/diagnóstico por imagen , Sustancia Negra/fisiopatología , Área Tegmental Ventral/diagnóstico por imagen , Área Tegmental Ventral/fisiopatología , Adulto Joven
4.
Hum Mol Genet ; 26(5): 888-900, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28069796

RESUMEN

Methylenetetrahydrofolate reductase (MTHFR) generates methyltetrahydrofolate for methylation reactions. Severe MTHFR deficiency results in homocystinuria and neurologic impairment. Mild MTHFR deficiency (677C > T polymorphism) increases risk for complex traits, including neuropsychiatric disorders. Although low dietary folate impacts brain development, recent concerns have focused on high folate intake following food fortification and increased vitamin use. Our goal was to determine whether high dietary folate during pregnancy affects brain development in murine offspring. Female mice were placed on control diet (CD) or folic acid-supplemented diet (FASD) throughout mating, pregnancy and lactation. Three-week-old male pups were evaluated for motor and cognitive function. Tissues from E17.5 embryos, pups and dams were collected for choline/methyl metabolite measurements, immunoblotting or gene expression of relevant enzymes. Brains were examined for morphology of hippocampus and cortex. Pups of FASD mothers displayed short-term memory impairment, decreased hippocampal size and decreased thickness of the dentate gyrus. MTHFR protein levels were reduced in FASD pup livers, with lower concentrations of phosphocholine and glycerophosphocholine in liver and hippocampus, respectively. FASD pup brains showed evidence of altered acetylcholine availability and Dnmt3a mRNA was reduced in cortex and hippocampus. E17.5 embryos and placentas from FASD dams were smaller. MTHFR protein and mRNA were reduced in embryonic liver, with lower concentrations of choline, betaine and phosphocholine. Embryonic brain displayed altered development of cortical layers. In summary, high folate intake during pregnancy leads to pseudo-MTHFR deficiency, disturbed choline/methyl metabolism, embryonic growth delay and memory impairment in offspring. These findings highlight the unintended negative consequences of supplemental folic acid.


Asunto(s)
Ácido Fólico/efectos adversos , Homocistinuria/genética , Memoria a Corto Plazo/efectos de los fármacos , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Espasticidad Muscular/genética , Acetilcolina/genética , Acetilcolina/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Dieta/efectos adversos , Femenino , Ácido Fólico/administración & dosificación , Homocistinuria/inducido químicamente , Homocistinuria/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/fisiopatología , Metilación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Ratones , Espasticidad Muscular/inducido químicamente , Espasticidad Muscular/patología , Embarazo , Trastornos Psicóticos/genética , Trastornos Psicóticos/patología
5.
Neuroimage ; 118: 397-405, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26004502

RESUMEN

The myelin g-ratio, defined as the ratio between the inner and the outer diameter of the myelin sheath, is a fundamental property of white matter that can be computed from a simple formula relating the myelin volume fraction to the fiber volume fraction or the axon volume fraction. In this paper, a unique combination of magnetization transfer, diffusion imaging and histology is presented, providing a novel method for in vivo magnetic resonance imaging of the axon volume fraction and the myelin g-ratio. Our method was demonstrated in the corpus callosum of one cynomolgus macaque, and applied to obtain full-brain g-ratio maps in one healthy human subject and one multiple sclerosis patient. In the macaque, the g-ratio was relatively constant across the corpus callosum, as measured by both MRI and electron microscopy. In the human subjects, the g-ratio in multiple sclerosis lesions was higher than in normal appearing white matter, which was in turn higher than in healthy white matter. Measuring the g-ratio brings us one step closer to fully characterizing white matter non-invasively, making it possible to perform in vivo histology of the human brain during development, aging, disease and treatment.


Asunto(s)
Axones/ultraestructura , Encéfalo/ultraestructura , Imagen de Difusión por Resonancia Magnética/métodos , Vaina de Mielina/ultraestructura , Adulto , Animales , Cuerpo Calloso/ultraestructura , Humanos , Macaca fascicularis , Fenómenos Magnéticos , Masculino , Ratones Mutantes Neurológicos , Esclerosis Múltiple/patología
6.
Biochem J ; 461(2): 205-12, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24800750

RESUMEN

Hyperhomocysteinaemia can contribute to cognitive impairment and brain atrophy. MTRR (methionine synthase reductase) activates methionine synthase, which catalyses homocysteine remethylation to methionine. Severe MTRR deficiency results in homocystinuria with cognitive and motor impairments. An MTRR polymorphism may influence homocysteine levels and reproductive outcomes. The goal of the present study was to determine whether mild hyperhomocysteinaemia affects neurological function in a mouse model with Mtrr deficiency. Mtrr+/+, Mtrr+/gt and Mtrrgt/gt mice (3 months old) were assessed for short-term memory, brain volumes and hippocampal morphology. We also measured DNA methylation, apoptosis, neurogenesis, choline metabolites and expression of ChAT (choline acetyltransferase) and AChE (acetylcholinesterase) in the hippocampus. Mtrrgt/gt mice exhibited short-term memory impairment on two tasks. They had global DNA hypomethylation and decreased choline, betaine and acetylcholine levels. Expression of ChAT and AChE was increased and decreased respectively. At 3 weeks of age, they showed increased neurogenesis. In the cerebellum, mutant mice had DNA hypomethylation, decreased choline and increased expression of ChAT. Our work demonstrates that mild hyperhomocysteinaemia is associated with memory impairment. We propose a mechanism whereby a deficiency in methionine synthesis leads to hypomethylation and compensatory disturbances in choline metabolism in the hippocampus. This disturbance affects the levels of acetylcholine, a critical neurotransmitter in learning and memory.


Asunto(s)
Cerebelo/metabolismo , Ferredoxina-NADP Reductasa/genética , Hipocampo/metabolismo , Homocistinuria/metabolismo , Hiperhomocisteinemia/metabolismo , Memoria a Corto Plazo , Acetilcolinesterasa/genética , Acetilcolinesterasa/metabolismo , Animales , Apoptosis , Betaína/metabolismo , Cerebelo/patología , Colina/metabolismo , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Metilación de ADN , Modelos Animales de Enfermedad , Ferredoxina-NADP Reductasa/deficiencia , Expresión Génica , Hipocampo/patología , Homocisteína/metabolismo , Homocistinuria/genética , Homocistinuria/patología , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Masculino , Metionina/metabolismo , Ratones , Ratones Noqueados , Transmisión Sináptica
7.
Neuroimage ; 91: 344-52, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24462776

RESUMEN

Persistent pain is a central characteristic of neuropathic pain conditions in humans. Knowing whether rodent models of neuropathic pain produce persistent pain is therefore crucial to their translational applicability. We investigated the spared nerve injury (SNI) model of neuropathic pain and the formalin pain model in rats using positron emission tomography (PET) with the metabolic tracer [18F]fluorodeoxyglucose (FDG) to determine if there is ongoing brain activity suggestive of persistent pain. For the formalin model, under brief anesthesia we injected one hindpaw with 5% formalin and the FDG tracer into a tail vein. We then allowed the animals to awaken and observed pain behavior for 30min during the FDG uptake period. The rat was then anesthetized and placed in the scanner for static image acquisition, which took place between minutes 45 and 75 post-tracer injection. A single reference rat brain magnetic resonance image (MRI) was used to align the PET images with the Paxinos and Watson rat brain atlas. Increased glucose metabolism was observed in the somatosensory region associated with the injection site (S1 hindlimb contralateral), S1 jaw/upper lip and cingulate cortex. Decreases were observed in the prelimbic cortex and hippocampus. Second, SNI rats were scanned 3weeks post-surgery using the same scanning paradigm, and region-of-interest analyses revealed increased metabolic activity in the contralateral S1 hindlimb. Finally, a second cohort of SNI rats was scanned while anesthetized during the tracer uptake period, and the S1 hindlimb increase was not observed. Increased brain activity in the somatosensory cortex of SNI rats resembled the activity produced with the injection of formalin, suggesting that the SNI model may produce persistent pain. The lack of increased activity in S1 hindlimb with general anesthetic demonstrates that this effect can be blocked, as well as highlights the importance of investigating brain activity in awake and behaving rodents.


Asunto(s)
Química Encefálica/fisiología , Encéfalo/diagnóstico por imagen , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Neuralgia/metabolismo , Neuralgia/fisiopatología , Animales , Conducta Animal/fisiología , Dolor Crónico/diagnóstico por imagen , Fluorodesoxiglucosa F18 , Procesamiento de Imagen Asistido por Computador , Ligadura , Masculino , Neuralgia/diagnóstico por imagen , Dimensión del Dolor , Neuropatías Peroneas/diagnóstico por imagen , Neuropatías Peroneas/metabolismo , Neuropatías Peroneas/fisiopatología , Tomografía de Emisión de Positrones , Radiofármacos , Ratas , Ratas Sprague-Dawley , Neuropatía Tibial/diagnóstico por imagen , Neuropatía Tibial/metabolismo , Neuropatía Tibial/fisiopatología
8.
Ann Neurol ; 74(5): 709-20, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23818336

RESUMEN

OBJECTIVE: To define the functional significance of increased miR-155 expression in myeloid cells in multiple sclerosis (MS). METHODS: miR-155 expression levels were measured in CD14+ monocytes from untreated relapsing-remitting MS patients and compared to healthy controls. Similar microRNA (miRNA) analyses were performed in laser-captured CD68+ cells from perivascular (blood-derived macrophages) and parenchymal (microglia) brain regions in both active MS lesions and noninflammatory cases. Using human adult blood-derived macrophages and brain-derived microglia, in vitro experiments were performed to demonstrate how miR-155 influences the polarization state, phenotype, and functional properties of myeloid cells, in addition to their ability to subsequently impact adaptive T-cell responses. RESULTS: In MS, miR-155 expression was significantly increased in both peripheral circulating CD14+ monocytes and active lesions (CD68+ cells) compared to control donor monocytes and parenchymal microglia, respectively. In vitro, miR-155 was significantly increased in both M1-polarized primary human macrophages and microglia. Transfection of an miR-155 mimic increased proinflammatory cytokine secretion and costimulatory surface marker expression in both cell types; an miR-155 inhibitor decreased proinflammatory cytokine expression. Coculture experiments demonstrated that allogeneic T-cell responses were significantly enhanced in the presence of miR-155-transfected myeloid cells compared to controls. INTERPRETATION: Our results demonstrate that miR-155 regulates proinflammatory responses in both blood-derived and central nervous system (CNS)-resident myeloid cells, in addition to impacting subsequent adaptive immune responses. Differential miRNA expression may therefore provide insight into mechanisms responsible for distinct phenotypic and functional properties of myeloid cells, thus impacting their ability to influence CNS injury and repair.


Asunto(s)
Polaridad Celular/fisiología , MicroARNs/genética , Esclerosis Múltiple/genética , Células Mieloides/patología , Inmunidad Adaptativa , Adulto , Anciano , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Polaridad Celular/inmunología , Proliferación Celular , Femenino , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , MicroARNs/metabolismo , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
9.
Neurology ; 102(5): e209137, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38315945

RESUMEN

BACKGROUND AND OBJECTIVES: Sensitive, reliable, and scalable biomarkers are needed to accelerate the development of therapies for Parkinson disease (PD). In this study, we evaluate the biomarkers of early PD diagnosis, disease progression, and treatment effect collected in the SPARK. METHODS: Cinpanemab is a human-derived monoclonal antibody binding preferentially to aggregated forms of extracellular α-synuclein. SPARK was a randomized, double-blind, placebo-controlled, phase 2 multicenter trial evaluating 3 cinpanemab doses administered intravenously every 4 weeks for 52 weeks with an active treatment dose-blind extension period for up to 112 weeks. SPARK enrolled 357 participants diagnosed with PD within 3 years, aged 40-80 years, ≤2.5 on the modified Hoehn and Yahr scale, and with evidence of striatal dopaminergic deficit. The primary outcome was change from baseline in the Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale total score. Secondary and exploratory biomarker outcomes evaluated change from baseline at week 52 relative to placebo. Dopamine transporter SPECT and MRI were used to quantify changes in the nigrostriatal dopamine pathway and regional atrophy. CSF and plasma samples were used to assess change in total α-synuclein levels, α-synuclein seeding, and neurofilament light chain levels. SPARK was conducted from January 2018 to April 2021 and terminated due to lack of efficacy. RESULTS: Approximately 3.8% (15/398) of SPECT-imaged participants did not have evidence of dopaminergic deficit and were screen-failed. Binary classification of α-synuclein seeding designated 93% (110/118) of the enrolled CSF subgroup as positive for α-synuclein seeds at baseline. Clinical disease progression was observed, with no statistically significant difference in cinpanemab groups compared with that in placebo. Ninety-nine percent of participants with positive α-synuclein seeding remained positive through week 52. No statistically significant changes from baseline were observed between treatment groups and placebo across biomarker measures. Broadly, there was minimal annual change with high interindividual variability across biomarkers-with striatal binding ratios of the ipsilateral putamen showing the greatest mean change/SD over time. DISCUSSION: Biomarker results indicated enrollment of the intended population with early PD, but there was no significant correlation with disease progression or clear evidence of a cinpanemab treatment effect on biomarker measures. Suitable biomarkers for evaluating disease severity and progression in early PD trials are still needed. TRIAL REGISTRATION INFORMATION: NCT03318523 (clinicaltrials.gov/ct2/show/NCT03318523); Submitted October 24, 2017; First patient enrolled January 2018.


Asunto(s)
Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/diagnóstico por imagen , Enfermedad de Parkinson/tratamiento farmacológico , alfa-Sinucleína , Antiparkinsonianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Dopamina/metabolismo , Biomarcadores , Progresión de la Enfermedad , Método Doble Ciego
10.
Neurobiol Dis ; 54: 59-67, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454197

RESUMEN

Magnetic resonance imaging (MRI) studies have identified aberrant cortical structure in Alzheimer's disease (AD). The association between MRI-derived cortical morphometry measures and ß-amyloid, however, remains poorly understood. In this study, we explored the potential relationship between early alterations in cortical thickness and later stage ß-amyloid deposition, using a novel approach, in a transgenic AD mouse model. We acquired longitudinal anatomical MRI scans from mutant amyloid precursor protein (APP) transgenic mice and age-matched wild-type mice at 1 and 3.5months-of-age, and employed fully-automated image processing methods to derive objective, quantitative measures of cortical thickness on a region-of-interest basis. We also generated 3D quantitative immunohistochemistry (qIHC) volumes of deposited ß-amyloid burden from 18month-old transgenic mice using an automated, production-level process. These studies revealed thinner cortex in most regions in the 1month-old transgenic mice relative to age-matched wild-types, with the exception of the frontal, perirhinal/entorhinal, posterior cingulate, and retrosplenial cortical regions. Between 1 and 3.5months-of-age, the transgenic mice demonstrated stable or increasing cortical thickness, while the wild-type mice showed cortical thinning. Based on data from co-registered 3D MRI and qIHC volumes, we identified an association between abnormal, early, regional cortical thickness change over 2.5months and later ß-amyloid deposition. These observations suggest that the spatio-temporal pattern of early (pre-plaque) alterations in cerebral cortical structure is indicative of regional predisposition to later ß-amyloid pathology in a transgenic AD mouse model.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/patología , Procesamiento de Imagen Asistido por Computador/métodos , Enfermedad de Alzheimer/metabolismo , Animales , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos
11.
bioRxiv ; 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37808643

RESUMEN

Several PET studies have explored the relationship between ß-amyloid load and tau uptake at the early stages of Alzheimer's disease (AD) progression. Most of these studies have focused on the linear relationship between ß-amyloid and tau at the local level and their synergistic effect on different AD biomarkers. We hypothesize that patterns of spatial association between ß-amyloid and tau might be uncovered using alternative association metrics that account for linear as well as more complex, possible nonlinear dependencies. In the present study, we propose a new Canonical Distance Correlation Analysis (CDCA) to generate distinctive spatial patterns of the cross-correlation structure between tau, as measured by [18F]flortaucipir PET, and ß-amyloid, as measured by [18F]florbetapir PET, from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We found that the CDCA-based ß-amyloid scores were not only maximally distance-correlated to tau in cognitively normal (CN) controls and mild cognitive impairment (MCI), but also differentiated between low and high levels of ß-amyloid uptake. The most distinctive spatial association pattern was characterized by a spread of ß-amyloid covering large areas of the cortex and localized tau in the entorhinal cortex. More importantly, this spatial dependency varies according to cognition, which cannot be explained by the uptake differences in ß-amyloid or tau between CN and MCI subjects. Hence, the CDCA-based scores might be more accurate than the amyloid or tau SUVR for the enrollment in clinical trials of those individuals on the path of cognitive deterioration.

12.
Mol Genet Metab ; 106(2): 149-59, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22521626

RESUMEN

The brain is particularly sensitive to folate metabolic disturbances, since methyl groups are critical for its functions. Methylenetetrahydrofolate reductase (MTHFR) generates the primary circulatory form of folate required for homocysteine remethylation to methionine. Neurological disturbances have been described in homocystinuria caused by severe MTHFR deficiency. The goal of this study was to determine if behavioral anomalies are present in severe Mthfr-deficient (Mthfr(-/-)) mice and to identify neurobiological changes that could contribute to these anomalies. Adult male mice of 3 Mthfr genotypes (+/+, +/-, -/-) were tested on motor, anxiety, exploratory and cognitive tasks. Volumes (whole brain and hippocampus) and morphology, global DNA methylation, apoptosis, expression of choline acetyltransferase (ChAT) and glucocorticoid receptor (GR), and concentrations of choline metabolites were assessed in hippocampus. Mthfr(-/-) mice had impairments in motor function and in short- and long-term memory, increased exploratory behavior and decreased anxiety. They showed decreased whole brain and hippocampal volumes, reduced thickness of the pyramidal cell layer of CA1 and CA3, and increased apoptosis in hippocampus. There was a disturbance in choline metabolism as manifested by differences in acetylcholine, betaine or glycerophosphocholine concentrations, and by increased ChAT levels. Mthfr(-/-) mice also had increased GR mRNA and protein. Our study has revealed significant anomalies in affective behavior and impairments in memory of Mthfr(-/-) mice. We identified structural changes, increased apoptosis, altered choline metabolism and GR dysregulation in hippocampus. These findings, as well as some similar observations in cerebellum, could contribute to the behavioral changes and suggest that choline is a critical metabolite in homocystinuria.


Asunto(s)
Hipocampo/metabolismo , Hipocampo/patología , Homocistinuria/diagnóstico , Espasticidad Muscular/diagnóstico , Animales , Apoptosis/genética , Conducta Animal , Encéfalo/metabolismo , Encéfalo/patología , Cerebelo/metabolismo , Cerebelo/patología , Colina/metabolismo , Colina O-Acetiltransferasa/metabolismo , Metilación de ADN , Homocisteína/sangre , Homocistinuria/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Ratones , Ratones Noqueados , Espasticidad Muscular/genética , Tamaño de los Órganos , Trastornos Psicóticos/diagnóstico , Trastornos Psicóticos/genética , Receptores de Glucocorticoides/metabolismo
13.
Alzheimer Dis Assoc Disord ; 26(1): 8-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22354138

RESUMEN

BACKGROUND: Florbetapir F 18 (F-AV-45) is a positron emission tomography imaging ligand for the detection of amyloid aggregation associated with Alzheimer disease. Earlier data showed that florbetapir F 18 binds with high affinity to ß-amyloid (Aß) plaques in human brain homogenates (Kd=3.7 nM) and has favorable imaging pharmacokinetic properties, including rapid brain penetration and washout. This study used human autopsy brain tissue to evaluate the correlation between in vitro florbetapir F 18 binding and Aß density measured by established neuropathologic methods. METHODS: The localization and density of florbetapir F 18 binding in frozen and formalin-fixed paraffin-embedded sections of postmortem brain tissue from 40 patients with a varying degree of neurodegenerative pathology was assessed by standard florbetapir F 18 autoradiography and correlated with the localization and density of Aß identified by silver staining, thioflavin S staining, and immunohistochemistry. RESULTS: There were strong quantitative correlations between florbetapir F 18 tissue binding and both Aß plaques identified by light microscopy (Silver staining and thioflavin S fluorescence) and by immunohistochemical measurements of Aß using 3 antibodies recognizing different epitopes of the Aß peptide. Florbetapir F 18 did not bind to neurofibrillary tangles. CONCLUSIONS: Florbetapir F 18 selectively binds Aß in human brain tissue. The binding intensity was quantitatively correlated with the density of Aß plaques identified by standard neuropathologic techniques and correlated with the density of Aß measured by immunohistochemistry. As Aß plaques are a defining neuropathologic feature for Alzheimer disease, these results support the use of florbetapir F 18 as an amyloid positron emission tomography ligand to identify the presence of Alzheimer disease pathology in patients with signs and symptoms of progressive late-life cognitive impairment.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Compuestos de Anilina/análisis , Encéfalo/diagnóstico por imagen , Glicoles de Etileno/análisis , Placa Amiloide/diagnóstico por imagen , Radiofármacos/análisis , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/análisis , Compuestos de Anilina/química , Autopsia , Autorradiografía , Encéfalo/metabolismo , Encéfalo/patología , Glicoles de Etileno/química , Humanos , Ovillos Neurofibrilares/diagnóstico por imagen , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Tomografía de Emisión de Positrones
14.
JAMA ; 305(3): 275-83, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21245183

RESUMEN

CONTEXT: The ability to identify and quantify brain ß-amyloid could increase the accuracy of a clinical diagnosis of Alzheimer disease. OBJECTIVE: To determine if florbetapir F 18 positron emission tomographic (PET) imaging performed during life accurately predicts the presence of ß-amyloid in the brain at autopsy. DESIGN, SETTING, AND PARTICIPANTS: Prospective clinical evaluation conducted February 2009 through March 2010 of florbetapir-PET imaging performed on 35 patients from hospice, long-term care, and community health care facilities near the end of their lives (6 patients to establish the protocol and 29 to validate) compared with immunohistochemistry and silver stain measures of brain ß-amyloid after their death used as the reference standard. PET images were also obtained in 74 young individuals (18-50 years) presumed free of brain amyloid to better understand the frequency of a false-positive interpretation of a florbetapir-PET image. MAIN OUTCOME MEASURES: Correlation of florbetapir-PET image interpretation (based on the median of 3 nuclear medicine physicians' ratings) and semiautomated quantification of cortical retention with postmortem ß-amyloid burden, neuritic amyloid plaque density, and neuropathological diagnosis of Alzheimer disease in the first 35 participants autopsied (out of 152 individuals enrolled in the PET pathological correlation study). RESULTS: Florbetapir-PET imaging was performed a mean of 99 days (range, 1-377 days) before death for the 29 individuals in the primary analysis cohort. Fifteen of the 29 individuals (51.7%) met pathological criteria for Alzheimer disease. Both visual interpretation of the florbetapir-PET images and mean quantitative estimates of cortical uptake were correlated with presence and quantity of ß-amyloid pathology at autopsy as measured by immunohistochemistry (Bonferroni ρ, 0.78 [95% confidence interval, 0.58-0.89]; P <.001]) and silver stain neuritic plaque score (Bonferroni ρ, 0.71 [95% confidence interval, 0.47-0.86]; P <.001). Florbetapir-PET images and postmortem results rated as positive or negative for ß-amyloid agreed in 96% of the 29 individuals in the primary analysis cohort. The florbetapir-PET image was rated as amyloid negative in the 74 younger individuals in the nonautopsy cohort. CONCLUSIONS: Florbetapir-PET imaging was correlated with the presence and density of ß-amyloid. These data provide evidence that a molecular imaging procedure can identify ß-amyloid pathology in the brains of individuals during life. Additional studies are required to understand the appropriate use of florbetapir-PET imaging in the clinical diagnosis of Alzheimer disease and for the prediction of progression to dementia.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/análisis , Compuestos de Anilina , Química Encefálica , Glicoles de Etileno , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Anciano , Autopsia , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Reacciones Falso Positivas , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Estudios Prospectivos
15.
Am J Pathol ; 174(5): 1880-90, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349355

RESUMEN

Remyelination of lesions in the central nervous system contributes to neural repair following clinical relapses in multiple sclerosis. Remyelination is initiated by recruitment and differentiation of oligodendrocyte progenitor cells (OPCs) into myelinating oligodendrocytes. Simvastatin, a blood-brain barrier-permeable statin in multiple sclerosis clinical trials, has been shown to impact the in vitro processes that have been implicated in remyelination. Animals were fed a cuprizone-supplemented diet for 6 weeks to induce localized demyelination in the corpus callosum; subsequent return to normal diet for 3 weeks stimulated remyelination. Simvastatin was injected intraperitoneally during the period of coincident demyelination and OPC maturation (weeks 4 to 6), throughout the entire period of OPC responses (weeks 4 to 9), or during the remyelination-only phase (weeks 7 to 9). Simvastatin treatment (weeks 4 to 6) caused a decrease in myelin load and both Olig2(strong) and Nkx2.2(strong) OPC numbers. Simvastatin treatment (weeks 4 to 9 and 7 to 9) caused a decrease in myelin load, which was correlated with a reduction in Nkx2.2(strong) OPCs and an increase in Olig2(strong) cells, suggesting that OPCs were maintained in an immature state (Olig2(strong)/Nkx2.2(weak)). NogoA+ oligodendrocyte numbers were decreased during all simvastatin treatment regimens. Our findings suggest that simvastatin inhibits central nervous system remyelination by blocking progenitor differentiation, indicating the need to monitor effects of systemic immunotherapies that can access the central nervous system on brain tissue-repair processes.


Asunto(s)
Anticolesterolemiantes/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Simvastatina/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Western Blotting , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quelantes/administración & dosificación , Cuprizona/administración & dosificación , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/fisiología , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/patología , Proteínas del Tejido Nervioso/fisiología , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factores de Transcripción/fisiología , Proteínas de Pez Cebra
16.
J Alzheimers Dis ; 73(2): 543-557, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31796668

RESUMEN

BACKGROUND: Several positron emission tomography (PET) studies have explored the relationship between amyloid-ß (Aß), glucose metabolism, and the APOEɛ4 genotype. It has been reported that APOEɛ4, and not aggregated Aß, contributes to glucose hypometabolism in pre-clinical stages of Alzheimer's disease (AD) pathology. OBJECTIVE: We hypothesize that typical measurements of Aß taken either from composite regions-of-interest with relatively high burden actually cover significant patterns of the relationship with glucose metabolism. In contrast, spatially weighted measures of Aß are more related to glucose metabolism in cognitively normal (CN) aging and mild cognitive impairment (MCI). METHODS: We have generated a score of amyloid burden based on a joint singular value decomposition (SVD) of the cross-correlation structure between glucose metabolism, as measured by [18F]2-fluoro-2-deoxyglucose (FDG) PET, and Aß, as measured by [18F]florbetapir PET, from the Alzheimer's Disease Neuroimaging Initiative study. This SVD-based score reveals cortical regions where a reduced glucose metabolism is maximally correlated with distributed patterns of Aß. RESULTS: From an older population of CN and MCI subjects, we found that the SVD-based Aß score was significantly correlated with glucose metabolism in several cortical regions. Additionally, the corresponding Aß network has hubs that contribute to distributed glucose hypometabolism, which, in turn, are not necessarily foci of Aß deposition. CONCLUSIONS: Our approach uncovered hidden patterns of the glucose metabolism-Aß relationship. We showed that the SVD-based Aß score produces a stronger relationship with decreasing glucose metabolism than either APOEɛ4 genotype or global measures of Aß burden.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/metabolismo , Glucosa/metabolismo , Anciano , Anciano de 80 o más Años , Apolipoproteína E4/genética , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/metabolismo , Disfunción Cognitiva/diagnóstico por imagen , Femenino , Fluorodesoxiglucosa F18 , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Tomografía de Emisión de Positrones
17.
Int J Dev Neurosci ; 80(4): 257-266, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32115740

RESUMEN

BACKGROUND: There is currently no targeted treatment available for neonatal arterial ischemic strokes (NAIS). Epidemiological studies demonstrated that perinatal infection/inflammation, peripartum hypoxia, and occlusion of the internal carotid tree are the main determinants of NAIS. The well-established benefit of therapeutic hypothermia (HT) in neonatal encephalopathy due to diffuse hypoxia-ischemia provides a rationale for the potential use of HT as a neuroprotective strategy in NAIS. METHODS: We used a rat model to reproduce the most prevalent human physiopathological scenario of NAIS. The neuroprotective effect of HT was measured by morphometric magnetic resonance imaging, [18 F] fluorodeoxyglucose (FDG) metabolic activity by positron emission tomography/computed tomography, and behavioral tests. RESULTS: HT (a) prevented the occurrence of 44% of NAIS, (b) reduced the volume of strokes by 37%, (c) enhanced [18 F] FDG metabolic activity within the territory of the occluded carotid artery, and (d) improved motor behavior. Both morphometric and metabolic techniques showed consistently that HT provided a neuroprotective effect located in the motor cortex, hippocampus, and caudate-putamen. CONCLUSION: Through combining anatomical, metabolic imaging, and behavioral studies, our study provides evidence of neuroprotective effects of HT in NAIS. These results are potentially translational to human NAIS.


Asunto(s)
Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/terapia , Accidente Cerebrovascular Isquémico/prevención & control , Neuroprotección , Animales , Animales Recién Nacidos , Núcleo Caudado/diagnóstico por imagen , Femenino , Hipocampo/diagnóstico por imagen , Humanos , Hipoxia-Isquemia Encefálica/diagnóstico por imagen , Recién Nacido , Accidente Cerebrovascular Isquémico/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Actividad Motora , Corteza Motora/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones , Putamen/diagnóstico por imagen , Ratas , Ratas Endogámicas Lew
18.
J Nucl Med ; 60(1): 100-106, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29777003

RESUMEN

SUV ratios (SUVRs) are commonly used to quantify tracer uptake in amyloid-ß PET. Here, we explore the impact of target and reference region-of-interest (ROI) selection on SUVR effect sizes using interventional data from the ongoing phase 1b PRIME study (NCT01677572) of aducanumab (BIIB037) in patients with prodromal or mild Alzheimer disease. Methods: The florbetapir PET SUVR was calculated at baseline (screening) and at weeks 26 and 54 for patients randomized to receive placebo and each of 4 aducanumab doses (1, 3, 6, and 10 mg/kg) using the whole cerebellum, cerebellar gray matter, cerebellar white matter, pons, and subcortical white matter as reference regions. In addition to the prespecified composite cortex target ROI, individual cerebral cortical ROIs were assessed as targets. Results: Of the reference regions used, subcortical white matter, cerebellar white matter, and the pons, alone or in combination, generated the largest effect sizes. The use of the anterior cingulate cortex as a target ROI resulted in larger effect sizes than the use of the composite cortex. SUVR calculations were not affected by correction for brain volume changes over time. Conclusion: Dose- and time-dependent reductions in the amyloid PET SUVR were consistently observed with aducanumab only in cortical regions prone to amyloid plaque deposition, regardless of the reference region used. These data support the hypothesis that florbetapir SUVR responses associated with aducanumab treatment are a result of specific dose- and time-dependent reductions in the amyloid burden in patients with Alzheimer disease.


Asunto(s)
Amiloide/metabolismo , Anticuerpos Monoclonales Humanizados/metabolismo , Tomografía de Emisión de Positrones/normas , Adulto , Transporte Biológico , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Estándares de Referencia
19.
Mol Neurobiol ; 56(6): 4175-4191, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30288696

RESUMEN

Folate is an important B vitamin required for methylation reactions, nucleotide and neurotransmitter synthesis, and maintenance of homocysteine at nontoxic levels. Its metabolism is tightly linked to that of choline, a precursor to acetylcholine and membrane phospholipids. Low folate intake and genetic variants in folate metabolism, such as the methylenetetrahydrofolate reductase (MTHFR) 677 C>T polymorphism, have been suggested to impact brain function and increase the risk for cognitive decline and late-onset Alzheimer's disease. Our study aimed to assess the impact of genetic and nutritional disturbances in folate metabolism, and their potential interaction, on features of cognitive decline and brain biochemistry in a mouse model. Wild-type and Mthfr+/- mice, a model for the MTHFR 677 C>T polymorphism, were fed control or folate-deficient diets from weaning until 8 and 10 months of age. We observed short-term memory impairment measured by the novel object paradigm, altered transcriptional levels of synaptic markers and epigenetic enzymes, as well as impaired choline metabolism due to the Mthfr+/- genotype in cortex or hippocampus. We also detected changes in mRNA levels of Presenillin-1, neurotrophic factors, one-carbon metabolic and epigenetic enzymes, as well as reduced levels of S-adenosylmethionine and acetylcholine, due to the folate-deficient diet. These findings shed further insights into the mechanisms by which genetic and dietary folate metabolic disturbances increase the risk for cognitive decline and suggest that these mechanisms are distinct.


Asunto(s)
Envejecimiento/patología , Encéfalo/patología , Dieta , Ácido Fólico/metabolismo , Homocistinuria/complicaciones , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Espasticidad Muscular/complicaciones , Péptidos beta-Amiloides/metabolismo , Animales , Ansiedad/complicaciones , Ansiedad/fisiopatología , Encéfalo/fisiopatología , Supervivencia Celular , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Colina/metabolismo , Islas de CpG/genética , Metilación de ADN/genética , Epigénesis Genética , Ácido Glutámico/metabolismo , Homocistinuria/fisiopatología , Hígado/metabolismo , Masculino , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Memoria a Corto Plazo , Metilación , Ratones Endogámicos BALB C , Espasticidad Muscular/fisiopatología , Factores de Crecimiento Nervioso/metabolismo , Neuronas/patología , Fosfolípidos/metabolismo , Trastornos Psicóticos/complicaciones , Trastornos Psicóticos/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , S-Adenosilmetionina/metabolismo , Transmisión Sináptica
20.
J Neuropathol Exp Neurol ; 78(12): 1130-1146, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31665376

RESUMEN

Astrocytes are increasingly recognized as active contributors to the disease process in multiple sclerosis (MS), rather than being merely reactive. We investigated the expression of a selected microRNA (miRNA) panel that could contribute both to the injury and to the recovery phases of the disease. Individual astrocytes were laser microdissected from brain sections. We then compared the miRNAs' expressions in MS and control brain samples at different lesional stages in white versus grey matter regions. In active MS lesions, we found upregulation of ischemia-related miRNAs in white but not grey matter, often with reversion to the normal state in inactive lesions. In contrast to our previous findings on MS macrophages, expression of 2 classical inflammatory-related miRNAs, miRNA-155 and miRNA-146a, was reduced in astrocytes from active and chronic active MS lesions in white and grey matter, suggesting a lesser direct pathogenetic role for these miRNAs in astrocytes. miRNAs within the categories regulating aquaporin4 (-100, -145, -320) and glutamate transport/apoptosis/neuroprotection (-124a, -181a, and -29a) showed some contrasting responses. The regional and lesion-stage differences of expression of these miRNAs indicate the remarkable ability of astrocytes to show a wide range of selective responses in the face of differing insults and phases of resolution.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/patología , MicroARNs/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Encefalitis/complicaciones , Encefalitis/metabolismo , Femenino , Sustancia Gris/patología , Humanos , Masculino , Esclerosis Múltiple/etiología , Sustancia Blanca/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA