Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Psychiatry ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38871852

RESUMEN

The amyloid cascade hypothesis assumes that the development of Alzheimer's disease (AD) is driven by a self-perpetuating cycle, in which ß-amyloid (Aß) accumulation leads to Tau pathology and neuronal damages. A particular mutation (A673T) of the amyloid precursor protein (APP) was identified among Icelandic population. It provides a protective effect against Alzheimer- and age-related cognitive decline. This APP mutation leads to the reduced production of Aß with A2T (position in peptide sequence) change (Aßice). In addition, Aßice has the capacity to form protective heterodimers in association with wild-type Aß. Despite the emerging interest in Aßice during the last decade, the impact of Aßice on events associated with the amyloid cascade has never been reported. First, the effects of Aßice were evaluated in vitro by electrophysiology on hippocampal slices and by studying synapse morphology in cortical neurons. We showed that Aßice protects against endogenous Aß-mediated synaptotoxicity. Second, as several studies have outlined that a single intracerebral administration of Aß can worsen Aß deposition and cognitive functions several months after the inoculation, we evaluated in vivo the long-term effects of a single inoculation of Aßice or Aß-wild-type (Aßwt) in the hippocampus of transgenic mice (APPswe/PS1dE9) over-expressing Aß1-42 peptide. Interestingly, we found that the single intra-hippocampal inoculation of Aßice to mice rescued synaptic density and spatial memory losses four months post-inoculation, compared with Aßwt inoculation. Although Aß load was not modulated by Aßice infusion, the amount of Tau-positive neuritic plaques was significantly reduced. Finally, a lower phagocytosis by microglia of post-synaptic compounds was detected in Aßice-inoculated animals, which can partly explain the increased density of synapses in the Aßice animals. Thus, a single event as Aßice inoculation can improve the fate of AD-associated pathology and phenotype in mice several months after the event. These results open unexpected fields to develop innovative therapeutic strategies against AD.

2.
Brain ; 146(1): 149-166, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-35298632

RESUMEN

Huntington's disease is a fatal neurodegenerative disease characterized by striatal neurodegeneration, aggregation of mutant Huntingtin and the presence of reactive astrocytes. Astrocytes are important partners for neurons and engage in a specific reactive response in Huntington's disease that involves morphological, molecular and functional changes. How reactive astrocytes contribute to Huntington's disease is still an open question, especially because their reactive state is poorly reproduced in experimental mouse models. Here, we show that the JAK2-STAT3 pathway, a central cascade controlling astrocyte reactive response, is activated in the putamen of Huntington's disease patients. Selective activation of this cascade in astrocytes through viral gene transfer reduces the number and size of mutant Huntingtin aggregates in neurons and improves neuronal defects in two complementary mouse models of Huntington's disease. It also reduces striatal atrophy and increases glutamate levels, two central clinical outcomes measured by non-invasive magnetic resonance imaging. Moreover, astrocyte-specific transcriptomic analysis shows that activation of the JAK2-STAT3 pathway in astrocytes coordinates a transcriptional program that increases their intrinsic proteolytic capacity, through the lysosomal and ubiquitin-proteasome degradation systems. This pathway also enhances their production and exosomal release of the co-chaperone DNAJB1, which contributes to mutant Huntingtin clearance in neurons. Together, our results show that the JAK2-STAT3 pathway controls a beneficial proteostasis response in reactive astrocytes in Huntington's disease, which involves bi-directional signalling with neurons to reduce mutant Huntingtin aggregation, eventually improving disease outcomes.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Animales , Ratones , Enfermedad de Huntington/genética , Astrocitos/metabolismo , Proteostasis , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
3.
Neurobiol Dis ; 181: 106116, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37054900

RESUMEN

Tauopathy is a typical feature of Alzheimer's disease of major importance because it strongly correlates with the severity of cognitive deficits experienced by patients. During the pathology, it follows a characteristic spatiotemporal course which takes its origin in the transentorhinal cortex, and then gradually invades the entire forebrain. To study the mechanisms of tauopathy, and test new therapeutic strategies, it is necessary to set-up relevant and versatile in vivo models allowing to recapitulate tauopathy. With this in mind, we have developed a model of tauopathy by overexpression of the human wild-type Tau protein in retinal ganglion cells in mice (RGCs). This overexpression led to the presence of hyperphosphorylated forms of the protein in the transduced cells as well as to their progressive degeneration. The application of this model to mice deficient in TREM2 (Triggering Receptor Expressed on Myeloid cells-2, an important genetic risk factor for AD) as well as to 15-month-old mice showed that microglia actively participate in the degeneration of RGCs. Surprisingly, although we were able to detect the transgenic Tau protein up to the terminal arborization of RGCs at the level of the superior colliculi, spreading of the transgenic Tau protein to post-synaptic neurons was detected only in aged animals. This suggests that there may be neuron-intrinsic- or microenvironment mediators facilitating this spreading that appear with aging.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Modelos Animales de Enfermedad , Glicoproteínas de Membrana/metabolismo , Ratones Transgénicos , Microglía/metabolismo , Receptores Inmunológicos/metabolismo , Células Ganglionares de la Retina/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/patología , Vías Visuales/metabolismo
4.
Brain ; 144(4): 1167-1182, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33842937

RESUMEN

Deposits of different abnormal forms of tau in neurons and astrocytes represent key anatomo-pathological features of tauopathies. Although tau protein is highly enriched in neurons and poorly expressed by astrocytes, the origin of astrocytic tau is still elusive. Here, we used innovative gene transfer tools to model tauopathies in adult mouse brains and to investigate the origin of astrocytic tau. We showed in our adeno-associated virus (AAV)-based models and in Thy-Tau22 transgenic mice that astrocytic tau pathology can emerge secondarily to neuronal pathology. By designing an in vivo reporter system, we further demonstrated bidirectional exchanges of tau species between neurons and astrocytes. We then determined the consequences of tau accumulation in astrocytes on their survival in models displaying various status of tau aggregation. Using stereological counting of astrocytes, we report that, as for neurons, soluble tau species are highly toxic to some subpopulations of astrocytes in the hippocampus, whereas the accumulation of tau aggregates does not affect their survival. Thus, astrocytes are not mere bystanders of neuronal pathology. Our results strongly suggest that tau pathology in astrocytes may significantly contribute to clinical symptoms.


Asunto(s)
Astrocitos/patología , Hipocampo/patología , Tauopatías/patología , Proteínas tau/toxicidad , Animales , Humanos , Masculino , Ratones , Neuronas/patología , Agregado de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/toxicidad , Tauopatías/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
5.
Neurobiol Dis ; 155: 105398, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34019997

RESUMEN

The role played by microglia has taken the center of the stage in the etiology of Alzheimer's disease (AD). Several genome-wide association studies carried out on large cohorts of patients have indeed revealed a large number of genetic susceptibility factors corresponding to genes involved in neuroinflammation and expressed specifically by microglia in the brain. Among these genes TREM2, a cell surface receptor expressed by microglia, arouses strong interest because its R47H variant confers a risk of developing AD comparable to the ε4 allele of the APOE gene. Since this discovery, a growing number of studies have therefore examined the role played by TREM2 in the evolution of amyloid plaques and neurofibrillary tangles, the two brain lesions characteristic of AD. Many studies report conflicting results, reflecting the complex nature of microglial activation in AD. Here, we investigated the impact of TREM2 deficiency in the THY-Tau22 transgenic line, a well-characterized model of tauopathy. Our study reports an increase in the severity of tauopathy lesions in mice deficient in TREM2 occurring at an advanced stage of the pathology. This exacerbation of pathology was associated with a reduction in microglial activation indicated by typical morphological features and altered expression of specific markers. However, it was not accompanied by any further changes in memory performance. Our longitudinal study confirms that a defect in microglial TREM2 signaling leads to an increase in neuronal tauopathy occurring only at late stages of the disease.


Asunto(s)
Modelos Animales de Enfermedad , Glicoproteínas de Membrana/deficiencia , Microglía/metabolismo , Receptores Inmunológicos/deficiencia , Tauopatías/metabolismo , Antígenos Thy-1/genética , Proteínas tau/genética , Animales , Femenino , Humanos , Estudios Longitudinales , Masculino , Aprendizaje por Laberinto/fisiología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Receptores Inmunológicos/genética , Tauopatías/genética , Tauopatías/patología
6.
Brain ; 141(2): 535-549, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29253129

RESUMEN

Tauopathies are neurodegenerative diseases characterized by the aggregation of tau protein. These pathologies exhibit a wide variety of clinical and anatomo-pathological presentations, which may result from different pathological mechanisms. Although tau inclusions are a common feature in all these diseases, recent evidence instead implicates small oligomeric aggregates as drivers of tau-induced toxicity. Hence in vivo model systems displaying either soluble or fibrillary forms of wild-type or mutant tau are needed to better identify their respective pathological pathways. Here we used adeno-associated viruses to mediate gene transfer of human tau to the rat brain to develop models of pure tauopathies. Two different constructs were used, each giving rise to a specific phenotype developing in less than 3 months. First, hTAUWT overexpression led to a strong hyperphosphorylation of the protein, which was associated with neurotoxicity in the absence of any significant aggregation. In sharp contrast, its co-expression with the pro-aggregation peptide TauRD-ΔK280 in the hTAUProAggr group strongly promoted its aggregation into Gallyas-positive neurofibrillary tangles, while preserving neuronal survival. Our results support the hypothesis that soluble tau species are key players of tau-induced neurodegeneration.


Asunto(s)
Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Tinción con Nitrato de Plata , Tauopatías/diagnóstico por imagen , Transducción Genética , Vimentina/metabolismo , Proteínas tau/genética
7.
Neuroimage ; 162: 306-321, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28899745

RESUMEN

Because they bridge the genetic gap between rodents and humans, non-human primates (NHPs) play a major role in therapy development and evaluation for neurological disorders. However, translational research success from NHPs to patients requires an accurate phenotyping of the models. In patients, magnetic resonance imaging (MRI) combined with automated segmentation methods has offered the unique opportunity to assess in vivo brain morphological changes. Meanwhile, specific challenges caused by brain size and high field contrasts make existing algorithms hard to use routinely in NHPs. To tackle this issue, we propose a complete pipeline, Primatologist, for multi-region segmentation. Tissue segmentation is based on a modular statistical model that includes random field regularization, bias correction and denoising and is optimized by expectation-maximization. To deal with the broad variety of structures with different relaxing times at 7 T, images are segmented into 17 anatomical classes, including subcortical regions. Pre-processing steps insure a good initialization of the parameters and thus the robustness of the pipeline. It is validated on 10 T2-weighted MRIs of healthy macaque brains. Classification scores are compared with those of a non-linear atlas registration, and the impact of each module on classification scores is thoroughly evaluated.


Asunto(s)
Algoritmos , Encéfalo/anatomía & histología , Macaca/anatomía & histología , Neuroimagen/métodos , Programas Informáticos , Animales , Atlas como Asunto , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional/métodos , Imagen por Resonancia Magnética
8.
Neuroimage ; 146: 1025-1037, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27989846

RESUMEN

The external pallidum (GPe) is a component of the indirect pathway centrally placed in the basal ganglia. Studies already demonstrated that the pharmacological disinhibition of the sensorimotor, associative, and limbic GPe produced dyskinesia, hyperactivity, and compulsive behaviors, respectively. The aim of this study was to investigate the cortical regions altered by the disinhibition of each GPe functional territory. Thus, 5 macaques were injected with bicuculline in sensorimotor, associative, and limbic sites of the GPe producing dyskinesia, hyperactivity, and compulsive behaviors, and underwent in vivo positron tomography with 18F-2-fluoro-2-deoxy-D-glucose to identify cortical dysfunctions related to GPe disinhibition. Blood cortisol levels were also quantified as a biomarker of anxiety for each condition. Our results showed that pallidal bicuculline injections in anesthetized animals reproducibly modified the activity of specific ipsilateral and contralateral cortical areas depending on the pallidal territory targeted. Bicuculline injections in the limbic GPe led to increased ipsilateral activations in limbic cortical regions (anterior insula, amygdala, and hippocampus). Injections in the associative vs. sensorimotor GPe increased the activity in the ipsilateral midcingulate vs. somatosensory and parietal cortices. Moreover, bicuculline injections increased blood cortisol levels only in animals injected in their limbic GPe. These are the first functional results supporting the model of opened cortico-striato-thalamo-cortical loops where modifications in a functional pallidal territory can impact cortical activities of the same functional territory but also cortical activities of other functional territories. This highlights the importance of the GPe as a crucial node in the top-down control of the cortico-striato-thalamo-cortical circuits from the frontal cortex to influence the perception, attention, and emotional processes at downstream (or non-frontal) cortical levels. Finally, we showed the implication of the ventral pallidum with the amygdala and the insular cortex in a circuit related to aversive processing that should be crucial for the production of anxious disorders.


Asunto(s)
Conducta Animal , Encéfalo/metabolismo , Globo Pálido/metabolismo , Animales , Bicuculina/administración & dosificación , Encéfalo/efectos de los fármacos , Conducta Compulsiva/metabolismo , Discinesias/metabolismo , Fluorodesoxiglucosa F18 , Antagonistas de Receptores de GABA-A/administración & dosificación , Globo Pálido/efectos de los fármacos , Hipercinesia/metabolismo , Macaca fascicularis , Macaca mulatta , Tomografía de Emisión de Positrones
9.
J Neurosci ; 32(32): 10809-18, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22875916

RESUMEN

Astrocytes and microglia become reactive under most brain pathological conditions, making this neuroinflammation process a surrogate marker of neuronal dysfunction. Neuroinflammation is associated with increased levels of translocator protein 18 kDa (TSPO) and binding sites for TSPO ligands. Positron emission tomography (PET) imaging of TSPO is thus commonly used to monitor neuroinflammation in preclinical and clinical studies. It is widely considered that TSPO PET signal reveals reactive microglia, although a few studies suggested a potential contribution of reactive astrocytes. Because astrocytes and microglia play very different roles, it is crucial to determine whether reactive astrocytes can also overexpress TSPO and yield to a detectable TSPO PET signal in vivo. We used a model of selective astrocyte activation through lentiviral gene transfer of the cytokine ciliary neurotrophic factor (CNTF) into the rat striatum, in the absence of neurodegeneration. CNTF induced an extensive activation of astrocytes, which overexpressed GFAP and become hypertrophic, whereas microglia displayed minimal increase in reactive markers. Two TSPO radioligands, [(18)F]DPA-714 [N,N-diethyl-2-(2-(4-(2-[(18)F]fluoroethoxy)phenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl)acetamide] and [(11)C]SSR180575 (7-chloro-N,N-dimethyl-5-[(11)C]methyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide), showed a significant binding in the lenti-CNTF-injected striatum that was saturated and displaced by PK11195 [N-methyl-N-(1-methylpropyl)-1-(2-chlorophenyl)-isoquinoline-3-carboxamide]. The volume of radioligand binding matched the GFAP immunopositive volume. TSPO mRNA levels were significantly increased, and TSPO protein was overexpressed by CNTF-activated astrocytes. We show that reactive astrocytes overexpress TSPO, yielding to a significant and selective binding of TSPO radioligands. Therefore, caution must be used when interpreting TSPO PET imaging in animals or patients because reactive astrocytes can contribute to the signal in addition to reactive microglia.


Asunto(s)
Astrocitos/diagnóstico por imagen , Astrocitos/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Tomografía de Emisión de Positrones , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Acetamidas/farmacocinética , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígeno CD11b/metabolismo , Proteínas de Unión al Calcio/metabolismo , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/efectos de los fármacos , Fluorodesoxiglucosa F18/metabolismo , Vectores Genéticos/genética , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Indoles/farmacocinética , Imagen por Resonancia Magnética , Masculino , Proteínas de Microfilamentos/metabolismo , Unión Proteica/efectos de los fármacos , ARN Mensajero/metabolismo , Ensayo de Unión Radioligante , Radiofármacos/farmacocinética , Ratas , Ratas Sprague-Dawley
10.
Front Neurosci ; 17: 1230814, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38274499

RESUMEN

Conventional histology of the brain remains the gold standard in the analysis of animal models. In most biological studies, standard protocols usually involve producing a limited number of histological slices to be analyzed. These slices are often selected into a specific anatomical region of interest or around a specific pathological lesion. Due to the lack of automated solutions to analyze such single slices, neurobiologists perform the segmentation of anatomical regions manually most of the time. Because the task is long, tedious, and operator-dependent, we propose an automated atlas segmentation method called giRAff, which combines rigid and affine registrations and is suitable for conventional histological protocols involving any number of single slices from a given mouse brain. In particular, the method has been tested on several routine experimental protocols involving different anatomical regions of different sizes and for several brains. For a given set of single slices, the method can automatically identify the corresponding slices in the mouse Allen atlas template with good accuracy and segmentations comparable to those of an expert. This versatile and generic method allows the segmentation of any single slice without additional anatomical context in about 1 min. Basically, our proposed giRAff method is an easy-to-use, rapid, and automated atlas segmentation tool compliant with a wide variety of standard histological protocols.

11.
Acta Neuropathol Commun ; 11(1): 66, 2023 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-37087498

RESUMEN

Alzheimer's disease (AD) is characterized by intracerebral deposition of abnormal proteinaceous assemblies made of amyloid-ß (Aß) peptides or tau proteins. These peptides and proteins induce synaptic dysfunctions that are strongly correlated with cognitive decline. Intracerebral infusion of well-defined Aß seeds from non-mutated Aß1-40 or Aß1-42 peptides can increase Aß depositions several months after the infusion. Familial forms of AD are associated with mutations in the amyloid precursor protein (APP) that induce the production of Aß peptides with different structures. The Aß Osaka (Aßosa mutation (E693Δ)) is located within the Aß sequence and thus the Aßosa peptides have different structures and properties as compared to non-mutated Aß1-42 peptides (Aßwt). Here, we wondered if a single exposure to this mutated Aß can worsen AD pathology as well as downstream events including cognition, cerebral connectivity and synaptic health several months after the inoculation. To answer this question we inoculated Aß1-42-bearing Osaka mutation (Aßosa) in the dentate gyrus of APPswe/PS1dE9 mice at the age of two months. Their cognition and cerebral connectivity were analyzed at 4 months post-inoculation by behavioral evaluation and functional MRI. Aß pathology as well as synaptic density were evaluated by histology. The impact of Aßosa peptides on synaptic health was also measured on primary cortical neurons. Remarkably, the intracerebral administration of Aßosa induced cognitive and synaptic impairments as well as a reduction of functional connectivity between different brain regions, 4 months post-inoculation. It increased Aß plaque depositions and increased Aß oligomers. This is the first study showing that a single, sporadic event as Aßosa inoculation can worsen the fate of the pathology and clinical outcome several months after the event. It suggests that a single inoculation of Aß regulates a large cascade of events for a long time.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Ratones , Animales , Ratones Transgénicos , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Cognición , Mutación/genética , Modelos Animales de Enfermedad
12.
Proc Natl Acad Sci U S A ; 106(10): 3988-93, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19234118

RESUMEN

Neuroimaging methods have considerably developed over the last decades and offer various noninvasive approaches for measuring cerebral metabolic fluxes connected to energy metabolism, including PET and magnetic resonance spectroscopy (MRS). Among these methods, (31)P MRS has the particularity and advantage to directly measure cerebral ATP synthesis without injection of labeled precursor. However, this approach is methodologically challenging, and further validation studies are required to establish (31)P MRS as a robust method to measure brain energy synthesis. In the present study, we performed a multimodal imaging study based on the combination of 3 neuroimaging techniques, which allowed us to obtain an integrated picture of brain energy metabolism and, at the same time, to validate the saturation transfer (31)P MRS method as a quantitative measurement of brain ATP synthesis. A total of 29 imaging sessions were conducted to measure glucose consumption (CMRglc), TCA cycle flux (V(TCA)), and the rate of ATP synthesis (V(ATP)) in primate monkeys by using (18)F-FDG PET scan, indirect (13)C MRS, and saturation transfer (31)P MRS, respectively. These 3 complementary measurements were performed within the exact same area of the brain under identical physiological conditions, leading to: CMRglc = 0.27 +/- 0.07 micromol x g(-1) x min(-1), V(TCA) = 0.63 +/- 0.12 micromol x g(-1) x min(-1), and V(ATP) = 7.8 +/- 2.3 micromol x g(-1) x min(-1). The consistency of these 3 fluxes with literature and, more interestingly, one with each other, demonstrates the robustness of saturation transfer (31)P MRS for directly evaluating ATP synthesis in the living brain.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Encéfalo/fisiología , Metabolismo Energético/fisiología , Imagenología Tridimensional/métodos , Animales , Ciclo del Ácido Cítrico , Fluorodesoxiglucosa F18 , Glucosa/metabolismo , Haplorrinos , Espectroscopía de Resonancia Magnética , Masculino , Tomografía de Emisión de Positrones , Reproducibilidad de los Resultados
13.
Microsc Res Tech ; 85(11): 3541-3552, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35855638

RESUMEN

This article uses microscopy images obtained from diverse anatomical regions of macaque brain for neuron semantic segmentation. The complex structure of brain, the large intra-class staining intensity difference within neuron class, the small inter-class staining intensity difference between neuron and tissue class, and the unbalanced dataset increase the difficulty of neuron semantic segmentation. To address this problem, we propose a multiscale segmentation- and error-guided iterative convolutional neural network (MSEG-iCNN) to improve the semantic segmentation performance in major anatomical regions of the macaque brain. After evaluating microscopic images from 17 anatomical regions, the semantic segmentation performance of neurons is improved by 10.6%, 4.0%, 1.5%, and 1.2% compared with Random Forest, FCN-8s, U-Net, and UNet++, respectively. Especially for neurons with brighter staining intensity in the anatomical regions such as lateral geniculate, globus pallidus and hypothalamus, the performance is improved by 66.1%, 23.9%, 11.2%, and 6.7%, respectively. Experiments show that our proposed method can efficiently segment neurons with a wide range of staining intensities. The semantic segmentation results are of great significance and can be further used for neuron instance segmentation, morphological analysis and disease diagnosis. Cell segmentation plays a critical role in extracting cerebral information, such as cell counting, cell morphometry and distribution analysis. Accurate automated neuron segmentation is challenging due to the complex structure of brain, the large intra-class staining intensity difference within neuron class, the small inter-class staining intensity difference between neuron and tissue class, and the unbalanced dataset. The proposed multiscale segmentation- and error-guided iterative convolutional neural network (MSEG-iCNN) improve the segmentation performance in 17 major anatomical regions of the macaque brain. HIGHLIGHTS: Cell segmentation plays a critical role in extracting cerebral information, such as cell counting, cell morphometry and distribution analysis. Accurate automated neuron segmentation is challenging due to the complex structure of brain, the large intra-class staining intensity difference within neuron class, the small inter-class staining intensity difference between neuron and tissue class, and the unbalanced dataset. The proposed multiscale segmentation- and error-guided iterative convolutional neural network (MSEG-iCNN) improve the segmentation performance in 17 major anatomical regions of the macaque brain.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Redes Neurales de la Computación , Animales , Encéfalo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Macaca , Neuronas
14.
Acta Neuropathol Commun ; 10(1): 112, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35974399

RESUMEN

Alzheimer's disease (AD) is characterized by intracerebral accumulations of extracellular amyloid-ß (Aß) plaques and intracellular tau pathology that spread in the brain. Three types of tau lesions occur in the form of neuropil threads, neurofibrillary tangles, and neuritic plaques i.e. tau aggregates within neurites surrounding Aß deposits. The cascade of events linking these lesions and synaptic or memory impairments are still debated. Intracerebral infusion of human AD brain extracts in Aß plaque-bearing mice that do not overexpress pathological tau proteins induces tau pathologies following heterotopic seeding of mouse tau protein. There is however little information regarding the downstream events including synaptic or cognitive repercussions of tau pathology induction in these models. In the present study, human AD brain extracts (ADbe) and control-brain extracts (Ctrlbe) were infused into the hippocampus of Aß plaque-bearing APPswe/PS1dE9 mice. Memory, synaptic density, as well as Aß plaque and tau aggregate loads, microgliosis, astrogliosis at the inoculation site and in connected regions (perirhinal/entorhinal cortex) were evaluated 4 and 8 months post-inoculation. ADbe inoculation produced the following effects: (i) memory deficit; (ii) increased Aß plaque deposition in proximity to the inoculation site; (iii) tau pathology induction; (iv) appearance of neuropil threads and neurofibrillary tangles next to the inoculation site with a spreading to connected regions. Neuritic plaque pathology was detected in both ADbe- and Ctrlbe-inoculated animals but ADbe inoculation increased the severity close to and at distance of the inoculation site. (v) Finally, ADbe inoculation reduced synaptic density in the vicinity to the inoculation site and in connected regions as the perirhinal/entorhinal cortex. Synaptic impairments were correlated with increased severity of neuritic plaques but not to other tau lesions or Aß lesions, suggesting that neuritic plaques are a culprit for synaptic loss. Synaptic density was also associated with microglial load.


Asunto(s)
Enfermedad de Alzheimer , Placa Amiloide , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Humanos , Ratones , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Proteínas tau/metabolismo
15.
Microsc Res Tech ; 84(10): 2311-2324, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33908123

RESUMEN

Accurate cerebral neuron segmentation is required before neuron counting and neuron morphological analysis. Numerous algorithms for neuron segmentation have been published, but they are mainly evaluated using limited subsets from a specific anatomical region, targeting neurons of clear contrast and/or neurons with similar staining intensity. It is thus unclear how these algorithms perform on cerebral neurons in diverse anatomical regions. In this article, we introduce and reliably evaluate existing machine learning algorithms using a data set of microscopy images of macaque brain. This data set highlights various anatomical regions (e.g., cortex, caudate, thalamus, claustrum, putamen, hippocampus, subiculum, lateral geniculate, globus pallidus, etc.), poor contrast, and staining intensity differences of neurons. The evaluation was performed using 10 architectures of six classic machine learning algorithms in terms of typical Recall, Precision, F-score, aggregated Jaccard index (AJI), as well as a performance ranking of algorithms. F-score of most of the algorithms is superior to 0.7. Deep learning algorithms facilitate generally higher F-scores. U-net with suitable layer depth has been evaluated to be excellent classifiers with F-score of 0.846 and 0.837 when performing cross validation. The evaluation and analysis indicate the performance gap among algorithms in various anatomical regions and the strengths and limitations of each algorithm. The comparative result highlights at the same time the importance and difficulty of neuron segmentation and provides clues for future improvement. To the best of our knowledge, this work is the first comprehensive study for neuron segmentation in such large-scale anatomical regions. Neuron segmentation plays a critical role in extracting cerebral information, such as neuron counting and neuron morphological analysis. Accurate automated cerebral neuron segmentation is a challenging task due to different kinds, poor contrast, staining intensity differences, and fuzzy boundaries of neurons. The comprehensive evaluation and analysis of performance among existing machine learning algorithms in diverse anatomical regions allows to make clear of the strengths and limitations of state-of-the-art algorithm. The comprehensive study provides clues for future improvement and creation of automated methods.


Asunto(s)
Algoritmos , Macaca , Animales , Encéfalo , Procesamiento de Imagen Asistido por Computador , Aprendizaje Automático , Neuronas
16.
Acta Neuropathol Commun ; 9(1): 165, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34641980

RESUMEN

Amyloid-ß (Aß) pathology transmission has been described in patients following iatrogenic exposure to compounds contaminated with Aß proteins. It can induce cerebral Aß angiopathy resulting in brain hemorrhages and devastating clinical impacts. Iatrogenic transmission of tau pathology is also suspected but not experimentally proven. In both scenarios, lesions were detected several decades after the putatively triggering medico-surgical act. There is however little information regarding the cognitive repercussions in individuals who do not develop cerebral hemorrhages. In the current study, we inoculated the posterior cingulate cortex and underlying corpus callosum of young adult primates (Microcebus murinus) with either Alzheimer's disease or control brain extracts. This led to widespread Aß and tau pathologies in all of the Alzheimer-inoculated animals following a 21-month-long incubation period (n = 12) whereas none of the control brain extract-inoculated animals developed such lesions (n = 6). Aß deposition affected almost all cortical regions. Tau pathology was also detected in Aß-deposit-free regions distant from the inoculation sites (e.g. in the entorhinal cortex), while some regions adjacent, but not connected, to the inoculation sites were spared (e.g. the occipital cortex). Alzheimer-inoculated animals developed cognitive deficits and cerebral atrophy compared to controls. These pathologies were induced using two different batches of Alzheimer brain extracts. This is the first experimental demonstration that tau can be transmitted by human brain extracts inoculations in a primate. We also showed for the first time that the transmission of widespread Aß and tau pathologies can be associated with cognitive decline. Our results thus reinforce the need to organize a systematic monitoring of individuals who underwent procedures associated with a risk of Aß and tau iatrogenic transmission. They also provide support for Alzheimer brain-inoculated primates as relevant models of Alzheimer pathology.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva , Proteínas tau/toxicidad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Cheirogaleidae , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Humanos , Enfermedad Iatrogénica
17.
Sci Total Environ ; 790: 148125, 2021 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380275

RESUMEN

Massive proliferation of some toxic marine dinoflagellates is responsible for the occurrence of harmful algal blooms and the contamination of fish and shellfish worldwide. Pinnatoxins (PnTx) (A-H) comprise an emerging phycotoxin family belonging to the cyclic imine toxin group. Interest has been focused on these lipophilic, fast-acting and highly potent toxins because they are widely found in contaminated shellfish, and can represent a risk for seafood consumers. PnTx display a potent antagonist effect on nicotinic acetylcholine receptors (nAChR), and in this study we assessed in vivo the ability of PnTx-G to cross physiological barriers to reach its molecular target. Radiolabeled [3H]-PnTx-G synthesized with good radiochemical purity and yield retained the high affinity of the natural toxin. Oral gavage or intravenous administration to adult rats and digital autoradiographic analyses revealed the biodistribution and toxicokinetics of [3H]-PnTx-G, which is rapidly cleared from blood, and accumulates in the liver and small intestine. The labeling of peripheral and brain adult/embryo rat tissues highlights its ability to cross the intestinal, blood-brain and placental barriers. High-resolution 3D-imaging and in vitro competition studies on rat embryo sections revealed the specificity of [3H]-PnTx-G binding and its selectivity for muscle and neuronal nAChR subtypes (such as α7 subtype). The use of a human perfused cotyledon model and mass spectrometry analyses disclosed that PnTx-G crosses the human placental barrier. The increasing worldwide occurrence of both the dinoflagellate Vulcanodinium rugosum and PnTx-contaminated shellfish, due to climate warming, raises concerns about the potential adverse impact that exposure to pinnatoxins may have for human health.


Asunto(s)
Placenta , Mariscos , Animales , Encéfalo , Femenino , Humanos , Embarazo , Ratas , Alimentos Marinos , Distribución Tisular
18.
Neuroimage ; 51(2): 586-98, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20206704

RESUMEN

Biomarkers and technologies similar to those used in humans are essential for the follow-up of Alzheimer's disease (AD) animal models, particularly for the clarification of mechanisms and the screening and validation of new candidate treatments. In humans, changes in brain metabolism can be detected by 1-deoxy-2-[(18)F] fluoro-D-glucose PET (FDG-PET) and assessed in a user-independent manner with dedicated software, such as Statistical Parametric Mapping (SPM). FDG-PET can be carried out in small animals, but its resolution is low as compared to the size of rodent brain structures. In mouse models of AD, changes in cerebral glucose utilization are usually detected by [(14)C]-2-deoxyglucose (2DG) autoradiography, but this requires prior manual outlining of regions of interest (ROI) on selected sections. Here, we evaluate the feasibility of applying the SPM method to 3D autoradiographic data sets mapping brain metabolic activity in a transgenic mouse model of AD. We report the preliminary results obtained with 4 APP/PS1 (64+/-1 weeks) and 3 PS1 (65+/-2 weeks) mice. We also describe new procedures for the acquisition and use of "blockface" photographs and provide the first demonstration of their value for the 3D reconstruction and spatial normalization of post mortem mouse brain volumes. Despite this limited sample size, our results appear to be meaningful, consistent, and more comprehensive than findings from previously published studies based on conventional ROI-based methods. The establishment of statistical significance at the voxel level, rather than with a user-defined ROI, makes it possible to detect more reliably subtle differences in geometrically complex regions, such as the hippocampus. Our approach is generic and could be easily applied to other biomarkers and extended to other species and applications.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Mapeo Encefálico/métodos , Encéfalo/diagnóstico por imagen , Glucosa/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Autorradiografía , Encéfalo/metabolismo , Radioisótopos de Carbono , Desoxiglucosa , Modelos Animales de Enfermedad , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Transgénicos , Tomografía de Emisión de Positrones , Presenilina-1/genética , Radiofármacos
19.
Acta Neuropathol Commun ; 8(1): 205, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33250056

RESUMEN

In humans, iatrogenic transmission of cerebral amyloid-ß (Aß)-amyloidosis is suspected following inoculation of pituitary-derived hormones or dural grafts presumably contaminated with Aß proteins as well as after cerebral surgeries. Experimentally, intracerebral inoculation of brain homogenate extracts containing misfolded Aß can seed Aß deposition in transgenic mouse models of amyloidosis or in non-human primates. The transmission of cerebral Aß is governed by the host and by the inoculated samples. It is critical to better characterize the propensities of different hosts to develop Aß deposition after contamination by an Aß-positive sample as well as to better assess which biological samples can transmit this lesion. Aß precursor protein (huAPPwt) mice express humanized non-mutated forms of Aß precursor protein and do not spontaneously develop Aß or amyloid deposits. We found that inoculation of Aß-positive brain extracts from Alzheimer patients in these mice leads to a sparse Aß deposition close to the alveus 18 months post-inoculation. However, it does not induce cortical or hippocampal Aß deposition. Secondary inoculation of apparently amyloid deposit-free hippocampal extracts from these huAPPwt mice to APPswe/PS1dE9 mouse models of amyloidosis enhanced Aß deposition in the alveus 9 months post-inoculation. This suggests that Aß seeds issued from human brain samples can persist in furtive forms in brain tissues while maintaining their ability to foster Aß deposition in receptive hosts that overexpress endogenous Aß. This work emphasizes the need for high-level preventive measures, especially in the context of neurosurgery, to prevent the risk of iatrogenic transmission of Aß lesions from samples with sparse amyloid markers.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloidosis/metabolismo , Encéfalo/metabolismo , Placa Amiloide/metabolismo , Extractos de Tejidos , Enfermedad de Alzheimer , Péptidos beta-Amiloides/administración & dosificación , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Hipocampo , Humanos , Enfermedad Iatrogénica , Inmunohistoquímica , Ratones , Ratones Transgénicos , Placa Amiloide/patología , Presenilina-1/genética
20.
J Cereb Blood Flow Metab ; 40(5): 1103-1116, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31238764

RESUMEN

The 18 kDa translocator protein (TSPO) is the main molecular target to image neuroinflammation by positron emission tomography (PET). However, TSPO-PET quantification is complex and none of the kinetic modelling approaches has been validated using a voxel-by-voxel comparison of TSPO-PET data with the actual TSPO levels of expression. Here, we present a single case study of binary classification of in vivo PET data to evaluate the statistical performance of different TSPO-PET quantification methods. To that end, we induced a localized and adjustable increase of TSPO levels in a non-human primate brain through a viral-vector strategy. We then performed a voxel-wise comparison of the different TSPO-PET quantification approaches providing parametric [18F]-DPA-714 PET images, with co-registered in vitro three-dimensional TSPO immunohistochemistry (3D-IHC) data. A data matrix was extracted from each brain hemisphere, containing the TSPO-IHC and TSPO-PET data for each voxel position. Each voxel was then classified as false or true, positive or negative after comparison of the TSPO-PET measure to the reference 3D-IHC method. Finally, receiver operating characteristic curves (ROC) were calculated for each TSPO-PET quantification method. Our results show that standard uptake value ratios using cerebellum as a reference region (SUVCBL) has the most optimal ROC score amongst all non-invasive approaches.


Asunto(s)
Encéfalo , Imagenología Tridimensional/métodos , Neuroimagen/métodos , Tomografía de Emisión de Positrones/métodos , Receptores de GABA/análisis , Animales , Radioisótopos de Flúor/análisis , Inmunohistoquímica , Macaca fascicularis , Masculino , Pirazoles/análisis , Pirimidinas/análisis , Radiofármacos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA