Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
BMJ Open ; 12(12): e067159, 2022 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-36585141

RESUMO

INTRODUCTION: The growing worldwide prevalence of Alzheimer's disease (AD) and the lack of effective treatments pose a dire medical challenge. Sleep disruption is also prevalent in the ageing population and is increasingly recognised as a risk factor and an early sign of AD. The ALFASleep project aims to characterise sleep with subjective and objective measurements in cognitively unimpaired middle/late middle-aged adults at increased risk of AD who are phenotyped with fluid and neuroimaging AD biomarkers. This will contribute to a better understanding of the pathophysiological mechanisms linking sleep with AD, thereby paving the way for the development of non-invasive biomarkers and preventive strategies targeting sleep. METHODS AND ANALYSIS: We will invite 200 participants enrolled in the ALFA+ (for ALzheimer and FAmilies) prospective observational study to join the ALFASleep study. ALFA+ participants are cognitively unimpaired middle-aged/late middle-aged adults who are followed up every 3 years with a comprehensive set of evaluations including neuropsychological tests, blood and cerebrospinal fluid (CSF) sampling, and MRI and positron emission tomography acquisition. ALFASleep participants will be additionally characterised with actigraphy and CSF-orexin-A measurements, and a subset (n=90) will undergo overnight polysomnography. We will test associations of sleep measurements and CSF-orexin-A with fluid biomarkers of AD and glial activation, neuroimaging outcomes and cognitive performance. In case we found any associations, we will test whether changes in AD and/or glial activation markers mediate the association between sleep and neuroimaging or cognitive outcomes and whether sleep mediates associations between CSF-orexin-A and AD biomarkers. ETHICS AND DISSEMINATION: The ALFASleep study protocol has been approved by the independent Ethics Committee Parc de Salut Mar, Barcelona (2018/8207/I). All participants have signed a written informed consent before their inclusion (approved by the same ethics committee). Study findings will be presented at national and international conferences and submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER: NCT04932473.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Pessoa de Meia-Idade , Doença de Alzheimer/diagnóstico , Biomarcadores , Cognição/fisiologia , Disfunção Cognitiva/diagnóstico , Estudos Observacionais como Assunto , Orexinas/líquido cefalorraquidiano , Qualidade do Sono
2.
J Med Chem ; 63(4): 1511-1525, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-31951127

RESUMO

We recently reported the discovery of a potent, selective, and brain-penetrant V1a receptor antagonist, which was not suitable for full development. Nevertheless, this compound was found to improve surrogates of social behavior in adults with autism spectrum disorder in an exploratory proof-of-mechanism study. Here we describe scaffold hopping that gave rise to triazolobenzodiazepines with improved pharmacokinetic properties. The key to balancing potency and selectivity while minimizing P-gp mediated efflux was fine-tuning of hydrogen bond acceptor basicity. Ascertaining a V1a antagonist specific brain activity pattern by pharmacological magnetic resonance imaging in the rat played a seminal role in guiding optimization efforts, culminating in the discovery of balovaptan (RG7314, RO5285119) 1. In a 12-week clinical phase 2 study in adults with autism spectrum disorder balovaptan demonstrated improvements in Vineland-II Adaptive Behavior Scales, a secondary end point comprising communication, socialization, and daily living skills. Balovaptan entered phase 3 clinical development in August 2018.


Assuntos
Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Transtorno do Espectro Autista/tratamento farmacológico , Benzodiazepinas/uso terapêutico , Piridinas/uso terapêutico , Receptores de Vasopressinas/metabolismo , Triazóis/uso terapêutico , Adolescente , Adulto , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/síntese química , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacocinética , Transtorno do Espectro Autista/metabolismo , Benzodiazepinas/síntese química , Benzodiazepinas/farmacocinética , Encéfalo/metabolismo , Criança , Ensaios Clínicos como Assunto , Descoberta de Drogas , Feminino , Humanos , Masculino , Mamíferos , Piridinas/síntese química , Piridinas/farmacocinética , Triazóis/síntese química , Triazóis/farmacocinética
3.
Ann Neurol ; 86(4): 561-571, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31359452

RESUMO

OBJECTIVE: Clinical trials targeting ß-amyloid peptides (Aß) for Alzheimer disease (AD) failed for arguable reasons that include selecting the wrong stages of AD pathophysiology or Aß being the wrong target. Targeting Aß to prevent cerebral amyloid angiopathy (CAA) has not been rigorously followed, although the causal role of Aß for CAA and related hemorrhages is undisputed. CAA occurs with normal aging and to various degrees in AD, where its impact and treatment is confounded by the presence of parenchymal Aß deposition. METHODS: APPDutch mice develop CAA in the absence of parenchymal amyloid, mimicking hereditary cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D). Mice were treated with a ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitor. We used 3-dimensional ultramicroscopy and immunoassays for visualizing CAA and assessing Aß in cerebrospinal fluid (CSF) and brain. RESULTS: CAA onset in mice was at 22 to 24 months, first in frontal leptomeningeal and superficial cortical vessels followed by vessels penetrating the cortical layers. CSF Aß increased with aging followed by a decrease of both Aß40 and Aß42 upon CAA onset, supporting the idea that combined reduction of CSF Aß40 and Aß42 is a specific biomarker for vascular amyloid. BACE1 inhibitor treatment starting at CAA onset and continuing for 4 months revealed a 90% Aß reduction in CSF and largely prevented CAA progression and associated pathologies. INTERPRETATION: This is the first study showing that Aß reduction at early disease time points largely prevents CAA in the absence of parenchymal amyloid. Our observation provides a preclinical basis for Aß-reducing treatments in patients at risk of CAA and in presymptomatic HCHWA-D. ANN NEUROL 2019;86:561-571.


Assuntos
Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Angiopatia Amiloide Cerebral/tratamento farmacológico , Progressão da Doença , Ácidos Picolínicos/uso terapêutico , Tiazinas/uso terapêutico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Biomarcadores/líquido cefalorraquidiano , Biomarcadores/metabolismo , Encéfalo/irrigação sanguínea , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Ácidos Picolínicos/farmacologia , Tiazinas/farmacologia
4.
Cell Rep ; 20(8): 1867-1880, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28834750

RESUMO

The NMDA receptor (NMDAR) antagonist ketamine elicits a long-lasting antidepressant response in patients with treatment-resistant depression. Understanding how antagonism of NMDARs alters synapse and circuit function is pivotal to developing circuit-based therapies for depression. Using virally induced gene deletion, ex vivo optogenetic-assisted circuit analysis, and in vivo chemogenetics and fMRI, we assessed the role of NMDARs in the medial prefrontal cortex (mPFC) in controlling depression-related behavior in mice. We demonstrate that post-developmental genetic deletion of the NMDAR subunit GluN2B from pyramidal neurons in the mPFC enhances connectivity between the mPFC and limbic thalamus, but not the ventral hippocampus, and reduces depression-like behavior. Using intersectional chemogenetics, we show that activation of this thalamocortical circuit is sufficient to elicit a decrease in despair-like behavior. Our findings reveal that GluN2B exerts input-specific control of pyramidal neuron innervation and identify a medial dorsal thalamus (MDT)→mPFC circuit that controls depression-like behavior.


Assuntos
Córtex Cerebral/metabolismo , Depressão/genética , Sinapses/metabolismo , Tálamo/metabolismo , Animais , Depressão/patologia , Camundongos
5.
EMBO J ; 36(13): 1837-1853, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28559417

RESUMO

Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) increase the risk for several neurodegenerative diseases including Alzheimer's disease and frontotemporal dementia (FTD). Homozygous TREM2 missense mutations, such as p.T66M, lead to the FTD-like syndrome, but how they cause pathology is unknown. Using CRISPR/Cas9 genome editing, we generated a knock-in mouse model for the disease-associated Trem2 p.T66M mutation. Consistent with a loss-of-function mutation, we observe an intracellular accumulation of immature mutant Trem2 and reduced generation of soluble Trem2 similar to patients with the homozygous p.T66M mutation. Trem2 p.T66M knock-in mice show delayed resolution of inflammation upon in vivo lipopolysaccharide stimulation and cultured macrophages display significantly reduced phagocytic activity. Immunohistochemistry together with in vivo TSPO small animal positron emission tomography (µPET) demonstrates an age-dependent reduction in microglial activity. Surprisingly, perfusion magnetic resonance imaging and FDG-µPET imaging reveal a significant reduction in cerebral blood flow and brain glucose metabolism. Thus, we demonstrate that a TREM2 loss-of-function mutation causes brain-wide metabolic alterations pointing toward a possible function of microglia in regulating brain glucose metabolism.


Assuntos
Encéfalo/patologia , Demência Frontotemporal/patologia , Glucose/metabolismo , Glicoproteínas de Membrana/genética , Microglia/fisiologia , Mutação de Sentido Incorreto , Perfusão , Receptores Imunológicos/genética , Animais , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Camundongos , Proteínas Mutantes/genética , Tomografia por Emissão de Pósitrons
6.
Sci Rep ; 6: 24523, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27080031

RESUMO

Functional magnetic resonance imaging (fMRI) has revolutionized neuroscience by opening a unique window that allows neurocircuitry function and pathological alterations to be probed non-invasively across brain disorders. Here we report a novel sustainable anesthesia procedure for small animal neuroimaging that overcomes shortcomings of anesthetics commonly used in rodent fMRI. The significantly improved preservation of cerebrovascular dynamics enhances sensitivity to neural activity changes for which it serves as a proxy in fMRI readouts. Excellent cross-species/strain applicability provides coherence among preclinical findings and is expected to improve translation to clinical fMRI investigations. The novel anesthesia procedure based on the GABAergic anesthetic etomidate was extensively validated in fMRI studies conducted in a range of genetically engineered rodent models of autism and strains commonly used for transgenic manipulations. Etomidate proved effective, yielded long-term stable physiology with basal cerebral blood flow of ~0.5 ml/g/min and full recovery. Cerebrovascular responsiveness of up to 180% was maintained as demonstrated with perfusion- and BOLD-based fMRI upon hypercapnic, pharmacological and sensory stimulation. Hence, etomidate lends itself as an anesthetic-of-choice for translational neuroimaging studies across rodent models of brain disorders.


Assuntos
Anestesia , Imageamento por Ressonância Magnética , Neuroimagem , Anestésicos Inalatórios/farmacologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Etomidato/farmacologia , Isoflurano/farmacologia , Medetomidina/farmacologia , Camundongos , Ratos , Especificidade da Espécie
7.
Neuroimage ; 112: 70-85, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25724758

RESUMO

Pharmacological magnetic resonance imaging (phMRI) of the brain has become a widely used tool in both preclinical and clinical drug research. One of its challenges is to condense the observed complex drug-induced brain-activation patterns into semantically meaningful metrics that can then serve as a basis for informed decision making. To aid interpretation of spatially distributed activation patterns, we propose here a set of multivariate metrics termed "domain gauges", which have been calibrated based on different classes of marketed or validated reference drugs. Each class represents a particular "domain" of interest, i.e., a specific therapeutic indication or mode of action. The drug class is empirically characterized by the unique activation pattern it evokes in the brain-the "domain profile". A domain gauge provides, for any tested intervention, a "classifier" as a measure of response strength with respect to the domain in question, and a "differentiator" as a measure of deviation from the domain profile, both along with error ranges. Capitalizing on our in-house database with an unprecedented wealth of standardized perfusion-based phMRI data obtained from rats subjected to various validated treatments, we exemplarily focused on 3 domains based on therapeutic indications: an antipsychotic, an antidepressant and an anxiolytic domain. The domain profiles identified as part of the gauge definition process, as well as the outputs of the gauges when applied to both reference and validation data, were evaluated for their reconcilability with prior biological knowledge and for their performance in drug characterization. The domain profiles provided quantitative activation patterns with high biological plausibility. The antipsychotic profile, for instance, comprised key areas (e.g., cingulate cortex, nucleus accumbens, ventral tegmental area, substantia nigra) which are believed to be strongly involved in mediating an antipsychotic effect, and which are in line with network-level dysfunctions observed in schizophrenia. The domain gauges plausibly positioned the vast majority of the pharmacological and even non-pharmacological treatments. The results also suggest the segregation of sub-domains based on, e.g., the mode of action. Upon judicious selection of domains and careful calibration of the gauges, our approach represents a valuable analytical tool for biological interpretation and decision making in drug discovery.


Assuntos
Encéfalo/efeitos dos fármacos , Imageamento por Ressonância Magnética/métodos , Psicotrópicos/farmacologia , Algoritmos , Animais , Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Antipsicóticos/farmacologia , Análise Discriminante , Masculino , Análise Multivariada , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
8.
PLoS One ; 9(9): e106156, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25181007

RESUMO

Translation of resting-state functional connectivity (FC) magnetic resonance imaging (rs-fMRI) applications from human to rodents has experienced growing interest, and bears a great potential in pre-clinical imaging as it enables assessing non-invasively the topological organization of complex FC networks (FCNs) in rodent models under normal and various pathophysiological conditions. However, to date, little is known about the organizational architecture of FCNs in rodents in a mentally healthy state, although an understanding of the same is of paramount importance before investigating networks under compromised states. In this study, we characterized the properties of resting-state FCN in an extensive number of Sprague-Dawley rats (n = 40) under medetomidine sedation by evaluating its modular organization and centrality of brain regions and tested for reproducibility. Fully-connected large-scale complex networks of positively and negatively weighted connections were constructed based on Pearson partial correlation analysis between the time courses of 36 brain regions encompassing almost the entire brain. Applying recently proposed complex network analysis measures, we show that the rat FCN exhibits a modular architecture, comprising six modules with a high between subject reproducibility. In addition, we identified network hubs with strong connections to diverse brain regions. Overall our results obtained under a straight medetomidine protocol show for the first time that the community structure of the rat brain is preserved under pharmacologically induced sedation with a network modularity contrasting from the one reported for deep anesthesia but closely resembles the organization described for the rat in conscious state.


Assuntos
Encéfalo/fisiologia , Sedação Profunda , Rede Nervosa/fisiologia , Animais , Imageamento por Ressonância Magnética , Masculino , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
9.
Neuroimage ; 74: 326-36, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23454471

RESUMO

Imaging methods that enable the investigation of functional networks both in human and animal brain provide important insights into mechanisms underlying pathologies including psychiatric disorders. Since the serotonergic receptor 1A (5-HT(1A)-R) has been strongly implicated in the pathophysiology of depressive and anxiety disorders, as well as in the action of antidepressant drugs, we investigated brain connectivity related to the 5-HT(1A)-R system by use of pharmacological functional magnetic resonance imaging in mice. We characterized functional connectivity elicited by activation of 5-HT(1A)-R and investigated how pharmacological and genetic manipulations of its function may modulate the evoked connectivity. Functional connectivity elicited by administration of the 5-HT(1A)-R agonist 8-OH-DPAT can be described by networks characterized by small-world attributes with nodes displaying highly concerted response patterns. Circuits identified comprised the brain structures known to be involved in stress-related disorders (e.g. prefrontal cortex, amygdala and hippocampus). The results also highlight the dorsomedial thalamus, a structure associated with fear processing, as a hub of the 5-HT(1A)-R functional network. Administration of a specific 5-HT(1A)-R antagonist or use of heterozygous 5-HT(1A)-R knockout mice significantly reduced functional connectivity elicited by 8-OH-DPAT. Whole brain functional connectivity analysis constitutes an attractive tool to characterize impairments in neurotransmission and the efficacy of pharmacological treatment in a comprehensive manner.


Assuntos
Mapeamento Encefálico/métodos , Encéfalo/metabolismo , Vias Neurais/fisiologia , Receptor 5-HT1A de Serotonina/genética , Receptor 5-HT1A de Serotonina/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Imageamento por Ressonância Magnética , Masculino , Transtornos Mentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/efeitos dos fármacos , Agonistas do Receptor de Serotonina/farmacologia
10.
Brain ; 135(Pt 11): 3265-81, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23169918

RESUMO

Whereas large injuries to the brain lead to considerable irreversible functional impairments, smaller strokes or traumatic lesions are often associated with good recovery. This recovery occurs spontaneously, and there is ample evidence from preclinical studies to suggest that adjacent undamaged areas (also known as peri-infarct regions) of the cortex 'take over' control of the disrupted functions. In rodents, sprouting of axons and dendrites has been observed in this region following stroke, while reduced inhibition from horizontal or callosal connections, or plastic changes in subcortical connections, could also occur. The exact mechanisms underlying functional recovery after small- to medium-sized strokes remain undetermined but are of utmost importance for understanding the human situation and for designing effective treatments and rehabilitation strategies. In the present study, we selectively destroyed large parts of the forelimb motor and premotor cortex of adult rats with an ischaemic injury. A behavioural test requiring highly skilled, cortically controlled forelimb movements showed that some animals recovered well from this lesion whereas others did not. To investigate the reasons behind these differences, we used anterograde and retrograde tracing techniques and intracortical microstimulation. Retrograde tracing from the cervical spinal cord showed a correlation between the number of cervically projecting corticospinal neurons present in the hindlimb sensory-motor cortex and good behavioural recovery. Anterograde tracing from the hindlimb sensory-motor cortex also showed a positive correlation between the degree of functional recovery and the sprouting of neurons from this region into the cervical spinal cord. Finally, intracortical microstimulation confirmed the positive correlation between rewiring of the hindlimb sensory-motor cortex and the degree of forelimb motor recovery. In conclusion, these experiments suggest that following stroke to the forelimb motor cortex, cells in the hindlimb sensory-motor area reorganize and become functionally connected to the cervical spinal cord. These new connections, probably in collaboration with surviving forelimb neurons and more complex indirect connections via the brainstem, play an important role for the recovery of cortically controlled behaviours like skilled forelimb reaching.


Assuntos
Membro Anterior/fisiopatologia , Membro Posterior/fisiopatologia , Córtex Motor/fisiologia , Tratos Piramidais/fisiologia , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Endotelina-1 , Feminino , Imageamento por Ressonância Magnética/métodos , Córtex Motor/patologia , Córtex Motor/fisiopatologia , Destreza Motora/fisiologia , Regeneração Nervosa/fisiologia , Vias Neurais/fisiologia , Técnicas de Rastreamento Neuroanatômico/métodos , Neuroimagem/métodos , Tratos Piramidais/patologia , Ratos , Ratos Long-Evans , Medula Espinal/patologia , Acidente Vascular Cerebral/patologia
11.
Neuroimage ; 59(2): 1028-36, 2012 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-21945466

RESUMO

Two general pathological processes contribute to multiple sclerosis (MS): acute inflammation and degeneration. While magnetic resonance imaging (MRI) is highly sensitive in detecting abnormalities related to acute inflammation both clinically and in animal models of experimental autoimmune encephalomyelitis (EAE), the correlation of these readouts with acute and future disabilities has been found rather weak. This illustrates the need for imaging techniques addressing neurodegenerative processes associated with MS. In the present work we evaluated the sensitivity of different MRI techniques (T(2) mapping, macrophage tracking based on labeling cells in vivo by ultrasmall particles of iron oxide (USPIO), diffusion tensor imaging (DTI) and magnetization transfer imaging (MTI)) to detect histopathological changes in a novel animal model making use of intrinsic, temporally and spatially controlled triggering of oligodendrocyte cell death. This mouse model allows studying the MRI signature associated to neurodegenerative processes of MS in the absence of adaptive inflammatory components that appear to be foremost in the EAE models. Our results revealed pronounced T(2) hyperintensities in brain stem and cerebellar structures, which we attribute to structural alteration of white matter by pronounced vacuolation. Brain areas were found devoid of significant macrophage infiltration in line with the absence of a peripheral inflammatory response. The significant decrease in diffusion anisotropy derived from DTI measures in these structures is mainly caused by a pronounced decrease in diffusivity parallel to the fiber indicative of axonal damage. Triggering of oligodendrocyte ablation did not translate into a significant increase in radial diffusivity. Only minor decreases in MT ratio have been observed, which is attributed to inefficient removal of myelin debris.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Imageamento por Ressonância Magnética/métodos , Esclerose Múltipla/patologia , Oligodendroglia/patologia , Animais , Apoptose , Rastreamento de Células/métodos , Humanos , Camundongos , Camundongos Transgênicos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
12.
Methods Mol Biol ; 711: 511-33, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21279620

RESUMO

One of the major neuropathological changes characteristic of Alzheimer's disease (AD) is deposits of beta-amyloid plaques and neurofibrillary tangles in neocortical and subcortical regions of the AD brain. The histochemical detection of these lesions in postmortem brain tissue is necessary for definitive diagnosis of AD. Methods for their in vivo detection would greatly aid the diagnosis of AD in early stages when neuronal loss and related functional impairment are still limited and would also open the opportunity for effective therapeutic interventions. Magnetic resonance imaging (MRI) theoretically provides the spatial resolution needed to resolve amyloid-ß plaques. Although currently limited for clinical applications due to unfavorable long acquisition times, MRI has been used to visualize Aß plaques in AD mouse models. The ability to detect amyloid-positive brain lesions in vivo using non-invasive imaging would allow to track disease progression and to monitor the efficacy of potential therapies in disease-modifying studies using transgenic models resembling AD pathology. Here, we provide MRI protocols for in vivo (mouse) and ex vivo (AD tissue samples) amyloid plaque imaging and the procedure for correlating these with thioflavin-S and iron-staining histology. Current challenges and limitations are discussed.


Assuntos
Doença de Alzheimer/complicações , Imageamento por Ressonância Magnética/métodos , Placa Amiloide/complicações , Placa Amiloide/diagnóstico , Doença de Alzheimer/diagnóstico , Animais , Encéfalo/patologia , Humanos , Camundongos , Perfusão , Fenótipo , Ondas de Rádio , Reprodutibilidade dos Testes , Técnicas Estereotáxicas
13.
J Neurosci ; 31(3): 1069-80, 2011 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-21248132

RESUMO

Loss of oligodendrocytes is a feature of many demyelinating diseases including multiple sclerosis. Here, we have established and characterized a novel model of genetically induced adult oligodendrocyte death. Specific primary loss of adult oligodendrocytes leads to a well defined and highly reproducible course of disease development that can be followed longitudinally by magnetic resonance imaging. Histological and ultrastructural analyses revealed progressive myelin vacuolation, in parallel to disease development that includes motor deficits, tremor, and ataxia. Myelin damage and clearance were associated with induction of oligodendrocyte precursor cell proliferation, albeit with some regional differences. Remyelination was present in the mildly affected corpus callosum. Consequences of acutely induced cell death of adult oligodendrocytes included secondary axonal damage. Microglia were activated in affected areas but without significant influx of B-cells, T-helper cells, or T-cytotoxic cells. Analysis of the model on a RAG-1 (recombination activating gene-1)-deficient background, lacking functional lymphocytes, did not change the observed disease and pathology compared with immune-competent mice. We conclude that this model provides the opportunity to study the consequences of adult oligodendrocyte death in the absence of primary axonal injury and reactive cells of the adaptive immune system. Our results indicate that if the blood-brain barrier is not disrupted, myelin debris is not removed efficiently, remyelination is impaired, and axonal integrity is compromised, likely as the result of myelin detachment. This model will allow the evaluation of strategies aimed at improving remyelination to foster axon protection.


Assuntos
Axônios/patologia , Morte Celular/genética , Corpo Caloso/patologia , Bainha de Mielina/patologia , Oligodendroglia/patologia , Animais , Axônios/metabolismo , Contagem de Células , Corpo Caloso/metabolismo , Progressão da Doença , Imunofluorescência , Imageamento por Ressonância Magnética , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Microscopia Eletrônica , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Teste de Desempenho do Rota-Rod
14.
Eur Neuropsychopharmacol ; 21(4): 344-53, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20656461

RESUMO

Visualization of brain activity in humans and animals using functional magnetic resonance imaging (fMRI) is an established method for translational neuropsychopharmacology. It is useful to study the activity of defined brain structures, however it requires further refinement to allow more specific cellular analyses, like for instance, the activity of selected pools of brain cells. Here, we investigated brain activity in serotonergic pathways in the adult mouse brain by using acute pharmacological challenge of 5-hydroxytryptamine (5-HT) 1A receptors. We show that administration of the 5-HT(1A) receptor agonist 8-OH-DPAT prompts a dose-dependent reduction in local cerebral blood volume (CBV) in brain areas rich in neurons expressing post-synaptic 5-HT(1A) receptor, including the prefrontal cortex, hippocampus and amygdalar nuclei. Region-specific inhibition of the response by co-injection of 8-OH-DPAT with the selective 5-HT(1A) receptor antagonist WAY-100635, or in 5-HT(1A) knock-out mice, suggests that 5-HT(1A) receptors are the primary targets of the agonist. Overall, the data demonstrate the feasibility of mapping region-specific serotonergic transmission in the adult mouse brain in vivo by non-invasive fMRI. The method opens novel perspectives for investigating 5-HT(1A) receptor functions in mouse models of human pathologies resulting from a dysfunction of the 5-HT(1A) receptor or the serotonergic system, including depression and anxiety.


Assuntos
Mapeamento Encefálico/métodos , Encéfalo/irrigação sanguínea , Encéfalo/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Fluxo Sanguíneo Regional/efeitos dos fármacos , 8-Hidroxi-2-(di-n-propilamino)tetralina/administração & dosagem , 8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Anatomia Transversal , Animais , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estudos de Viabilidade , Cinética , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Piperazinas/farmacologia , Piridinas/farmacologia , Receptor 5-HT1A de Serotonina/genética , Agonistas do Receptor 5-HT1 de Serotonina/administração & dosagem , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Antagonistas do Receptor 5-HT1 de Serotonina/farmacologia
15.
NMR Biomed ; 24(4): 439-46, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-22945293

RESUMO

Functional MRI (fMRI) based on the blood oxygen level-dependent (BOLD) contrast is widely used in preclinical neuroscience. The small dimensions of rodent brain place high demands on spatial resolution, and hence on the sensitivity of the fMRI experiment. This work investigates the performance of a 400-MHz cryogenic quadrature transceive radiofrequency probe (CryoProbe) with respect to the enhancement of the BOLD sensitivity. For this purpose, BOLD fMRI experiments were performed in mice during electrical forepaw stimulation using the CryoProbe and a conventional room temperature surface coil of comparable dimensions. Image signal-to-noise ratio (SNR) and temporal SNR were evaluated as quality measures for individual images and for fMRI time series of images, resulting in gains (mean ± standard deviation) with factors of 3.1 ± 0.7 and 1.8 ± 1.0 when comparing the CryoProbe and room temperature coil. The CryoProbe thermal shield temperature did not affect the noise characteristics, with temporal noise levels being 63 ± 16% of the corresponding room temperature value. However, a significant effect on BOLD amplitudes was found, which was attributed to temperature-dependent baseline cerebral blood volumes. Defined local thermal conditions were found to be a critical parameter for achieving an optimal and reproducible fMRI signal. In summary, the CryoProbe represents an attractive alternative for the enhancement of image SNR, temporal SNR and BOLD sensitivity in mouse fMRI experiments.


Assuntos
Temperatura Baixa , Membro Anterior/fisiologia , Imageamento por Ressonância Magnética/instrumentação , Imageamento por Ressonância Magnética/métodos , Oxigênio/sangue , Ondas de Rádio , Córtex Somatossensorial/fisiologia , Animais , Temperatura Corporal/fisiologia , Mapeamento Encefálico , Estimulação Elétrica , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Couro Cabeludo/fisiologia , Marcadores de Spin
16.
NMR Biomed ; 24(10): 1295-301, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22223365

RESUMO

Insulin resistance is a central feature of type II diabetes and is associated with alterations in skeletal muscle lipid metabolism, which manifest themselves, in part, in increased intramyocellular lipid (IMCL) accumulation. The objective of this study was to assess noninvasively the levels of IMCL longitudinally in the tibialis anterior muscle of Lep(ob) /Lep(ob) (ob/ob) mice, a genetic model of obesity and mild diabetes, and Lep(ob) /+ (ob/+) heterozygous control animals, using (1) H MRS at 9.4 T. The use of a cryogenic surface coil transceiver leads to significant increases in sensitivity. Method implementation included the assessment of the reproducibility and spatial heterogeneity of the IMCL signal and the determination of T(2) relaxation times, as IMCL levels were expressed relative to the total creatine signal, and therefore the signal ratios had to be corrected for differences in T(2) relaxation. IMCL levels were found to be significantly higher in ob/ob mice relative to ob/+ heterozygous control mice that do not develop disease. An increase in IMCL levels was observed for ob/ob mice until weeks 16/17; after this time point, IMCL levels decreased again, reaching final levels that were slightly higher than the initial values. These noninvasively detected alterations in skeletal muscle lipid metabolism in ob/ob mice were accompanied by a transient increase in plasma insulin concentrations. This study indicates that IMCL may be reliably assessed in mouse tibialis anterior muscle using a cryogenic surface coil, implying that (1) H MRS at 9.4 T represents a useful technology for the noninvasive measurement of changes in lipid metabolism in the skeletal muscle that accompany obesity.


Assuntos
Temperatura Baixa , Metabolismo dos Lipídeos , Espectroscopia de Ressonância Magnética/instrumentação , Espectroscopia de Ressonância Magnética/métodos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Envelhecimento/metabolismo , Animais , Peso Corporal , Creatina/metabolismo , Estudos de Viabilidade , Insulina/metabolismo , Masculino , Camundongos , Camundongos Obesos , Marcadores de Spin , Propriedades de Superfície
17.
Pain ; 151(3): 655-663, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20851520

RESUMO

Functional magnetic resonance imaging (fMRI) using the blood oxygen level-dependent (BOLD) contrast was used to study sensory processing in the brain of isoflurane-anesthetized mice. The use of a cryogenic surface coil in a small animal 9.4T system provided the sensitivity required for detection and quantitative analysis of hemodynamic changes caused by neural activity in the mouse brain in response to electrical forepaw stimulation at different amplitudes. A gradient echo-echo planar imaging (GE-EPI) sequence was used to acquire five coronal brain slices of 0.5mm thickness. BOLD signal changes were observed in primary and secondary somatosensory cortices, the thalamus and the insular cortex, important regions involved in sensory and nociceptive processing. Activation was observed consistently bilateral despite unilateral stimulation of the forepaw. The temporal BOLD profile was segregated into two signal components with different temporal characteristics. The maximum BOLD amplitude of both signal components correlated strongly with the stimulation amplitude. Analysis of the dynamic behavior of the somatosensory 'fast' BOLD component revealed a decreasing signal decay rate constant k(off) with increasing maximum BOLD amplitude (and stimulation amplitude). This study demonstrates the feasibility of a robust BOLD fMRI protocol to study nociceptive processing in isoflurane-anesthetized mice. The reliability of the method allows for detailed analysis of the temporal BOLD profile and for investigation of somatosensory and noxious signal processing in the brain, which is attractive for characterizing genetically engineered mouse models.


Assuntos
Encéfalo/fisiopatologia , Percepção da Dor/fisiologia , Dor/fisiopatologia , Animais , Autorradiografia , Mapeamento Encefálico , Feminino , Membro Anterior/fisiopatologia , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Camundongos , Estimulação Física
18.
Neurobiol Dis ; 40(1): 284-92, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20600914

RESUMO

Deposition of beta-amyloid along cerebral vessels is found in most patients suffering from Alzheimer's disease. The effects of cerebral amyloid angiopathy (CAA) on the function of cerebral blood vessels were analyzed applying cerebral blood volume (CBV)-based fMRI to transgenic arcA beta mice. In a cortical brain region of interest (ROI), displaying high CAA, arcA beta mice older than 16 months showed reduced response to the vasodilatory substance acetazolamide compared to age-matched wild-type animals, both with regard to rate (vascular reactivity) and extent of vasodilation (maximal vasodilation). In a subcortical ROI, displaying little CAA, no genotype-specific decrease was observed, but maximal vasodilation decreased with age in arcA beta and wild-types. These findings indicate that vascular beta-amyloid deposits reduce the capacity of cerebral blood vessels to dilate upon demand, supporting the hypothesis that vascular beta-amyloid contributes to hypoperfusion and neurological deficits observed in AD and CAA. High diagnostic accuracy of the combined readouts in detecting vascular dysfunction in arcA beta mice was found.


Assuntos
Acetazolamida , Envelhecimento/genética , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Angiopatia Amiloide Cerebral/genética , Artérias Cerebrais/metabolismo , Camundongos Transgênicos , Vasodilatadores , Envelhecimento/efeitos dos fármacos , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Angiopatia Amiloide Cerebral/diagnóstico , Angiopatia Amiloide Cerebral/metabolismo , Artérias Cerebrais/efeitos dos fármacos , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/genética , Modelos Animais de Doenças , Marcadores Genéticos , Humanos , Camundongos , Valor Preditivo dos Testes
19.
Nat Neurosci ; 13(1): 97-104, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20010824

RESUMO

Little is known about the functional role of axotomized cortical neurons that survive spinal cord injury. Large thoracic spinal cord injuries in adult rats result in impairments of hindlimb function. Using retrograde tracers, we found that axotomized corticospinal axons from the hindlimb sensorimotor cortex sprouted in the cervical spinal cord. Mapping of these neurons revealed the emergence of a new forelimb corticospinal projection from the rostral part of the former hindlimb cortex. Voltage-sensitive dye (VSD) imaging and blood-oxygen-level-dependent functional magnetic resonance imaging (BOLD fMRI) revealed a stable expansion of the forelimb sensory map, covering in particular the former hindlimb cortex containing the rewired neurons. Therefore, axotomized hindlimb corticospinal neurons can be incorporated into the sensorimotor circuits of the unaffected forelimb.


Assuntos
Mapeamento Encefálico , Membro Posterior/fisiopatologia , Regeneração Nervosa/fisiologia , Tratos Piramidais/patologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Animais , Axotomia/métodos , Comportamento Animal , Vértebras Cervicais , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes , Membro Anterior/fisiopatologia , Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Potenciais da Membrana/fisiologia , Córtex Motor/irrigação sanguínea , Córtex Motor/fisiopatologia , Oxigênio/sangue , Tratos Piramidais/irrigação sanguínea , Pirazóis , Ratos , Ratos Endogâmicos Lew , Estatísticas não Paramétricas , Tiazóis
20.
Eur J Neurosci ; 30(10): 1860-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19912336

RESUMO

Multimodal non-invasive neuroimaging in rodents constitutes an attractive tool for studying neurobiological processes in vivo. At present, imaging studies of brain anatomy and function as well as the investigation of structure-function relationships belong to the standard repertoire of neuroscientists. Molecular imaging adds a new perspective. The mapping of the receptor distribution and receptor occupancy can nowadays be complemented by specific readouts of receptor function either by visualizing the activity of signaling pathways or mapping the physiological consequences of receptor stimulation. Molecular information is obtained through the use of imaging probes that combine a target-specific ligand with a reporter moiety that generates a signal that can be detected from outside the body. For imaging probes targeting the central nervous system, penetration of the intact blood-brain barrier constitutes a major hurdle. Molecular imaging generates specific information and therefore has a large potential for disease phenotyping (diagnostics), therapy development and monitoring of treatment response. Molecular imaging is still in its infancy and major developments in imaging technology, probe design and data analysis are required in order to make an impact. Rodent molecular neuroimaging will play an important role in the development of these tools.


Assuntos
Mapeamento Encefálico , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Expressão Gênica/fisiologia , Imagem Molecular/métodos , Receptores de Superfície Celular/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos , Ligantes , Imageamento por Ressonância Magnética/métodos , Camundongos , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Roedores , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA