Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Hum Brain Mapp ; 45(1): e26551, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38063289

RESUMO

The interaction between cerebellum and cerebrum participates widely in function from motor processing to high-level cognitive and affective processing. Because of the motor symptom, idiopathic generalized epilepsy (IGE) patients with generalized tonic-clonic seizure have been recognized to associate with motor abnormalities, but the functional interaction in the cerebello-cerebral circuit is still poorly understood. Resting-state functional magnetic resonance imaging data were collected for 101 IGE patients and 106 healthy controls. The voxel-based functional connectivity (FC) between cerebral cortex and the cerebellum was contacted. The functional gradient and independent components analysis were applied to evaluate cerebello-cerebral functional integration on the voxel-based FC. Cerebellar motor components were further linked to cerebellar gradient. Results revealed cerebellar motor functional modules were closely related to cerebral motor components. The altered mapping of cerebral motor components to cerebellum was observed in motor module in patients with IGE. In addition, patients also showed compression in cerebello-cerebral functional gradient between motor and cognition modules. Interestingly, the contribution of the motor components to the gradient was unbalanced between bilateral primary sensorimotor components in patients: the increase was observed in cerebellar cognitive module for the dominant hemisphere primary sensorimotor, but the decrease was found in the cerebellar cognitive module for the nondominant hemisphere primary sensorimotor. The present findings suggest that the cerebral primary motor system affects the hierarchical architecture of cerebellum, and substantially contributes to the functional integration evidence to understand the motor functional abnormality in IGE patients.


Assuntos
Epilepsia Generalizada , Imageamento por Ressonância Magnética , Humanos , Vias Neurais , Mapeamento Encefálico/métodos , Epilepsia Generalizada/diagnóstico por imagem , Epilepsia Generalizada/patologia , Córtex Cerebral/diagnóstico por imagem , Cerebelo/diagnóstico por imagem , Imunoglobulina E
2.
Brain ; 145(5): 1785-1804, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34605898

RESUMO

Alzheimer's disease involves many neurobiological alterations from molecular to macroscopic spatial scales, but we currently lack integrative, mechanistic brain models characterizing how factors across different biological scales interact to cause clinical deterioration in a way that is subject-specific or personalized. As important signalling molecules and mediators of many neurobiological interactions, neurotransmitter receptors are promising candidates for identifying molecular mechanisms and drug targets in Alzheimer's disease. We present a neurotransmitter receptor-enriched multifactorial brain model, which integrates spatial distribution patterns of 15 neurotransmitter receptors from post-mortem autoradiography with multiple in vivo neuroimaging modalities (tau, amyloid-ß and glucose PET, and structural, functional and arterial spin labelling MRI) in a personalized, generative, whole-brain formulation. In a heterogeneous aged population (n = 423, ADNI data), models with personalized receptor-neuroimaging interactions showed a significant improvement over neuroimaging-only models, explaining about 70% (±20%) of the variance in longitudinal changes to the six neuroimaging modalities. In Alzheimer's disease patients (n = 25, ADNI data), receptor-imaging interactions explained up to 39.7% (P < 0.003, family-wise error-rate-corrected) of inter-individual variability in cognitive deterioration, via an axis primarily affecting executive function. Notably, based on their contribution to the clinical severity in Alzheimer's disease, we found significant functional alterations to glutamatergic interactions affecting tau accumulation and neural activity dysfunction and GABAergic interactions concurrently affecting neural activity dysfunction, amyloid and tau distributions, as well as significant cholinergic receptor effects on tau accumulation. Overall, GABAergic alterations had the largest effect on cognitive impairment (particularly executive function) in our Alzheimer's disease cohort (n = 25). Furthermore, we demonstrate the clinical applicability of this approach by characterizing subjects based on individualized 'fingerprints' of receptor alterations. This study introduces the first robust, data-driven framework for integrating several neurotransmitter receptors, multimodal neuroimaging and clinical data in a flexible and interpretable brain model. It enables further understanding of the mechanistic neuropathological basis of neurodegenerative progression and heterogeneity, and constitutes a promising step towards implementing personalized, neurotransmitter-based treatments.


Assuntos
Doença de Alzheimer , Encéfalo , Disfunção Cognitiva , Idoso , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Disfunção Cognitiva/patologia , Humanos , Imageamento por Ressonância Magnética/métodos , Neuroimagem/métodos , Tomografia por Emissão de Pósitrons/métodos , Receptores de Neurotransmissores , Proteínas tau/metabolismo
3.
Hum Brain Mapp ; 43(6): 1821-1835, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35118777

RESUMO

Frontotemporal dementia in genetic forms is highly heterogeneous and begins many years to prior symptom onset, complicating disease understanding and treatment development. Unifying methods to stage the disease during both the presymptomatic and symptomatic phases are needed for the development of clinical trials outcomes. Here we used the contrastive trajectory inference (cTI), an unsupervised machine learning algorithm that analyzes temporal patterns in high-dimensional large-scale population datasets to obtain individual scores of disease stage. We used cross-sectional MRI data (gray matter density, T1/T2 ratio as a proxy for myelin content, resting-state functional amplitude, gray matter fractional anisotropy, and mean diffusivity) from 383 gene carriers (269 presymptomatic and 115 symptomatic) and a control group of 253 noncarriers in the Genetic Frontotemporal Dementia Initiative. We compared the cTI-obtained disease scores to the estimated years to onset (age-mean age of onset in relatives), clinical, and neuropsychological test scores. The cTI based disease scores were correlated with all clinical and neuropsychological tests (measuring behavioral symptoms, attention, memory, language, and executive functions), with the highest contribution coming from mean diffusivity. Mean cTI scores were higher in the presymptomatic carriers than controls, indicating that the method may capture subtle pre-dementia cerebral changes, although this change was not replicated in a subset of subjects with complete data. This study provides a proof of concept that cTI can identify data-driven disease stages in a heterogeneous sample combining different mutations and disease stages of genetic FTD using only MRI metrics.


Assuntos
Demência Frontotemporal , Estudos Transversais , Demência Frontotemporal/diagnóstico por imagem , Demência Frontotemporal/genética , Demência Frontotemporal/psicologia , Heterozigoto , Humanos , Idioma , Imageamento por Ressonância Magnética
4.
Neuroimage ; 225: 117431, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33045336

RESUMO

The identification of community structure in graphs continues to attract great interest in several fields. Network neuroscience is particularly concerned with this problem considering the key roles communities play in brain processes and functionality. Most methods used for community detection in brain graphs are based on the maximization of a parameter-dependent modularity function that often obscures the physical meaning and hierarchical organization of the partitions of network nodes. In this work, we present a new method able to detect communities at different scales in a natural, unrestricted way. First, to obtain an estimation of the information flow in the network we release random walkers to freely move over it. The activity of the walkers is separated into oscillatory modes by using empirical mode decomposition. After grouping nodes by their co-occurrence at each time scale, k-modes clustering returns the desired partitions. Our algorithm was first tested on benchmark graphs with favorable performance. Next, it was applied to real and simulated anatomical and/or functional connectomes in the macaque and human brains. We found a clear hierarchical repertoire of community structures in both the anatomical and the functional networks. The observed partitions range from the evident division in two hemispheres -in which all processes are managed globally- to specialized communities seemingly shaped by physical proximity and shared function. Additionally, the spatial scales of a network's community structure (characterized by a measure we term within-communities path length) appear inversely proportional to the oscillatory modes' average frequencies. The proportionality constant may constitute a network-specific propagation velocity for the information flow. Our results stimulate the research of hierarchical community organization in terms of temporal scales of information flow in the brain network.


Assuntos
Encéfalo/fisiologia , Vias Neurais/fisiologia , Algoritmos , Análise por Conglomerados , Humanos
5.
Brain ; 143(2): 661-673, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31989163

RESUMO

Most prevalent neurodegenerative disorders take decades to develop and their early detection is challenged by confounding non-pathological ageing processes. For all neurodegenerative conditions, we continue to lack longitudinal gene expression data covering their large temporal evolution, which hinders the understanding of the underlying dynamic molecular mechanisms. Here, we overcome this key limitation by introducing a novel gene expression contrastive trajectory inference (GE-cTI) method that reveals enriched temporal patterns in a diseased population. Evaluated on 1969 subjects in the spectrum of late-onset Alzheimer's and Huntington's diseases (from ROSMAP, HBTRC and ADNI datasets), this unsupervised machine learning algorithm strongly predicts neuropathological severity (e.g. Braak, amyloid and Vonsattel stages). Furthermore, when applied to in vivo blood samples at baseline (ADNI), it significantly predicts clinical deterioration and conversion to advanced disease stages, supporting the identification of a minimally invasive (blood-based) tool for early clinical screening. This technique also allows the discovery of genes and molecular pathways, in both peripheral and brain tissues, that are highly predictive of disease evolution. Eighty-five to ninety per cent of the most predictive molecular pathways identified in the brain are also top predictors in the blood. These pathways support the importance of studying the peripheral-brain axis, providing further evidence for a key role of vascular structure/functioning and immune system response. The GE-cTI is a promising tool for revealing complex neuropathological mechanisms, with direct implications for implementing personalized dynamic treatments in neurology.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Deterioração Clínica , Regulação da Expressão Gênica , Doença de Huntington/patologia , Algoritmos , Progressão da Doença , Diagnóstico Precoce , Expressão Gênica/fisiologia , Humanos , Doença de Huntington/genética
6.
Hum Brain Mapp ; 40(2): 638-651, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30368979

RESUMO

Previous positron emission tomography (PET) studies have quantified filamentous tau pathology using regions-of-interest (ROIs) based on observations of the topographical distribution of neurofibrillary tangles in post-mortem tissue. However, such approaches may not take full advantage of information contained in neuroimaging data. The present study employs an unsupervised data-driven method to identify spatial patterns of tau-PET distribution, and to compare these patterns to previously published "pathology-driven" ROIs. Tau-PET patterns were identified from a discovery sample comprised of 123 normal controls and patients with mild cognitive impairment or Alzheimer's disease (AD) dementia from the Swedish BioFINDER cohort, who underwent [18 F]AV1451 PET scanning. Associations with cognition were tested in a separate sample of 90 individuals from ADNI. BioFINDER [18 F]AV1451 images were entered into a robust voxelwise stable clustering algorithm, which resulted in five clusters. Mean [18 F]AV1451 uptake in the data-driven clusters, and in 35 previously published pathology-driven ROIs, was extracted from ADNI [18 F]AV1451 scans. We performed linear models comparing [18 F]AV1451 signal across all 40 ROIs to tests of global cognition and episodic memory, adjusting for age, sex, and education. Two data-driven ROIs consistently demonstrated the strongest or near-strongest effect sizes across all cognitive tests. Inputting all regions plus demographics into a feature selection routine resulted in selection of two ROIs (one data-driven, one pathology-driven) and education, which together explained 28% of the variance of a global cognitive composite score. Our findings suggest that [18 F]AV1451-PET data naturally clusters into spatial patterns that are biologically meaningful and that may offer advantages as clinical tools.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Neuroimagem/métodos , Reconhecimento Automatizado de Padrão/métodos , Tomografia por Emissão de Pósitrons/métodos , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Carbolinas , Análise por Conglomerados , Estudos de Coortes , Conjuntos de Dados como Assunto , Feminino , Humanos , Masculino
7.
PLoS Comput Biol ; 14(5): e1006136, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29795548

RESUMO

Brain stimulation can modulate the activity of neural circuits impaired by Alzheimer's disease (AD), having promising clinical benefit. However, all individuals with the same condition currently receive identical brain stimulation, with limited theoretical basis for this generic approach. In this study, we introduce a control theory framework for obtaining exogenous signals that revert pathological electroencephalographic activity in AD at a minimal energetic cost, while reflecting patients' biological variability. We used anatomical networks obtained from diffusion magnetic resonance images acquired by the Alzheimer's Disease Neuroimaging Initiative (ADNI) as mediators for the interaction between Duffing oscillators. The nonlinear nature of the brain dynamics is preserved, given that we extend the so-called state-dependent Riccati equation control to reflect the stimulation objective in the high-dimensional neural system. By considering nonlinearities in our model, we identified regions for which control inputs fail to correct abnormal activity. There are changes to the way stimulated regions are ranked in terms of the energetic cost of controlling the entire network, from a linear to a nonlinear approach. We also found that limbic system and basal ganglia structures constitute the top target locations for stimulation in AD. Patients with highly integrated anatomical networks-namely, networks having low average shortest path length, high global efficiency-are the most suitable candidates for the propagation of stimuli and consequent success on the control task. Other diseases associated with alterations in brain dynamics and the self-control mechanisms of the brain can be addressed through our framework.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Encéfalo , Imagem de Difusão por Ressonância Magnética/métodos , Eletroencefalografia/métodos , Neuroimagem/métodos , Algoritmos , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Simulação por Computador , Humanos , Processamento de Imagem Assistida por Computador , Dinâmica não Linear , Processamento de Sinais Assistido por Computador
8.
Alzheimers Dement ; 15(7): 961-984, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31327392

RESUMO

The incidence of stroke and dementia are diverging across the world, rising for those in low- and middle-income countries and falling in those in high-income countries. This suggests that whatever factors cause these trends are potentially modifiable. At the population level, neurological disorders as a group account for the largest proportion of disability-adjusted life years globally (10%). Among neurological disorders, stroke (42%) and dementia (10%) dominate. Stroke and dementia confer risks for each other and share some of the same, largely modifiable, risk and protective factors. In principle, 90% of strokes and 35% of dementias have been estimated to be preventable. Because a stroke doubles the chance of developing dementia and stroke is more common than dementia, more than a third of dementias could be prevented by preventing stroke. Developments at the pathological, pathophysiological, and clinical level also point to new directions. Growing understanding of brain pathophysiology has unveiled the reciprocal interaction of cerebrovascular disease and neurodegeneration identifying new therapeutic targets to include protection of the endothelium, the blood-brain barrier, and other components of the neurovascular unit. In addition, targeting amyloid angiopathy aspects of inflammation and genetic manipulation hold new testable promise. In the meantime, accumulating evidence suggests that whole populations experiencing improved education, and lower vascular risk factor profiles (e.g., reduced prevalence of smoking) and vascular disease, including stroke, have better cognitive function and lower dementia rates. At the individual levels, trials have demonstrated that anticoagulation of atrial fibrillation can reduce the risk of dementia by 48% and that systolic blood pressure lower than 140 mmHg may be better for the brain. Based on these considerations, the World Stroke Organization has issued a proclamation, endorsed by all the major international organizations focused on global brain and cardiovascular health, calling for the joint prevention of stroke and dementia. This article summarizes the evidence for translation into action.


Assuntos
Fibrilação Atrial/diagnóstico , Encéfalo/fisiopatologia , Demência/prevenção & controle , Hipertensão/diagnóstico , Acidente Vascular Cerebral/prevenção & controle , Fibrilação Atrial/tratamento farmacológico , Barreira Hematoencefálica , Transtornos Cerebrovasculares/fisiopatologia , Demência/epidemiologia , Saúde Global , Humanos , Hipertensão/tratamento farmacológico , Incidência , Acidente Vascular Cerebral/epidemiologia
9.
Neuroimage ; 179: 40-50, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29894824

RESUMO

Personalized Medicine (PM) seeks to assist the patients according to their specific treatment needs and potential intervention responses. However, in the neurological context, this approach is limited by crucial methodological challenges, such as the requirement for an understanding of the causal disease mechanisms and the inability to predict the brain's response to therapeutic interventions. Here, we introduce and validate the concept of the personalized Therapeutic Intervention Fingerprint (pTIF), which predicts the effectiveness of potential interventions for controlling a patient's disease evolution. Each subject's pTIF can be inferred from multimodal longitudinal imaging (e.g. amyloid-ß, metabolic and tau PET; vascular, functional and structural MRI). We studied an aging population (N = 331) comprising cognitively normal and neurodegenerative patients, longitudinally scanned using six different neuroimaging modalities. We found that the resulting pTIF vastly outperforms cognitive and clinical evaluations on predicting individual variability in gene expression (GE) profiles. Furthermore, after regrouping the patients according to their predicted primary single-target interventions, we observed that these pTIF-based subgroups present distinctively altered molecular pathway signatures, supporting the across-population identification of dissimilar pathological stages, in active correspondence with different therapeutic needs. The results further evidence the imprecision of using broad clinical categories for understanding individual molecular alterations and selecting appropriate therapeutic needs. To our knowledge, this is the first study highlighting the direct link between multifactorial brain dynamics, predicted treatment responses, and molecular alterations at the patient level. Inspired by the principles of PM, the proposed pTIF framework is a promising step towards biomarker-driven assisted therapeutic interventions, with additional important implications for selective enrollment of patients in clinical trials.


Assuntos
Encéfalo/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos , Imagem Multimodal/métodos , Doenças Neurodegenerativas/diagnóstico por imagem , Neuroimagem/métodos , Medicina de Precisão/métodos , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Tomografia por Emissão de Pósitrons , Transcriptoma
10.
Neuroimage ; 152: 60-77, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28257929

RESUMO

Generative models focused on multifactorial causal mechanisms in brain disorders are scarce and generally based on limited data. Despite the biological importance of the multiple interacting processes, their effects remain poorly characterized from an integrative analytic perspective. Here, we propose a spatiotemporal multifactorial causal model (MCM) of brain (dis)organization and therapeutic intervention that accounts for local causal interactions, effects propagation via physical brain networks, cognitive alterations, and identification of optimum therapeutic interventions. In this article, we focus on describing the model and applying it at the population-based level for studying late onset Alzheimer's disease (LOAD). By interrelating six different neuroimaging modalities and cognitive measurements, this model accurately predicts spatiotemporal alterations in brain amyloid-ß (Aß) burden, glucose metabolism, vascular flow, resting state functional activity, structural properties, and cognitive integrity. The results suggest that a vascular dysregulation may be the most-likely initial pathologic event leading to LOAD. Nevertheless, they also suggest that LOAD it is not caused by a unique dominant biological factor (e.g. vascular or Aß) but by the complex interplay among multiple relevant direct interactions. Furthermore, using theoretical control analysis of the identified population-based multifactorial causal network, we show the crucial advantage of using combinatorial over single-target treatments, explain why one-target Aß based therapies might fail to improve clinical outcomes, and propose an efficiency ranking of possible LOAD interventions. Although still requiring further validation at the individual level, this work presents the first analytic framework for dynamic multifactorial brain (dis)organization that may explain both the pathologic evolution of progressive neurological disorders and operationalize the influence of multiple interventional strategies.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Encéfalo/patologia , Progressão da Doença , Modelos Neurológicos , Idoso , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/fisiopatologia , Encéfalo/irrigação sanguínea , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Mapeamento Encefálico/métodos , Feminino , Glucose/metabolismo , Humanos , Masculino , Vias Neurais/irrigação sanguínea , Vias Neurais/diagnóstico por imagem , Vias Neurais/metabolismo , Vias Neurais/patologia , Processamento de Sinais Assistido por Computador
11.
Eur J Neurosci ; 46(4): 1956-1967, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28512863

RESUMO

Sports-related concussions lead to persistent anomalies of the brain structure and function that interact with the effects of normal ageing. Although post-mortem investigations have proposed a bio-signature of remote concussions, there is still no clear in vivo signature. In the current study, we characterized white matter integrity in retired athletes with a history of remote concussions by conducting a full-brain, diffusion-based connectivity analysis. Next, we combined MRI diffusion markers with MR spectroscopic, MRI volumetric, neurobehavioral and genetic markers to identify a multidimensional in vivo signature of remote concussions. Machine learning classifiers trained to detect remote concussions using this signature achieved detection accuracies up to 90% (sensitivity: 93%, specificity: 87%). These automated classifiers identified white matter integrity as the hallmark of remote concussions and could provide, following further validation, a preliminary unbiased detection tool to help medical and legal experts rule out concussion history in patients presenting or complaining about late-life abnormal cognitive decline.


Assuntos
Concussão Encefálica/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Futebol Americano/lesões , Hóquei/lesões , Aprendizado de Máquina , Rede Nervosa/diagnóstico por imagem , Idoso , Traumatismos em Atletas/diagnóstico por imagem , Traumatismos em Atletas/psicologia , Concussão Encefálica/etiologia , Concussão Encefálica/psicologia , Futebol Americano/psicologia , Hóquei/psicologia , Humanos , Aprendizado de Máquina/tendências , Imageamento por Ressonância Magnética/tendências , Masculino , Pessoa de Meia-Idade , Esportes
12.
Curr Opin Neurol ; 30(6): 623-629, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29095718

RESUMO

PURPOSE OF REVIEW: This article provides a brief overview of relevant cerebrovascular mechanisms implicated in late-onset Alzheimer's disease (LOAD) development, and highlights the main reasons for incorporating novel cerebrovascular biomarkers to the models defining a multifactorial LOAD pathogenesis. We also discuss how novel brain mapping techniques and multifactorial data-driven models are having a critical role on understanding LOAD and may be particularly useful for identifying effective therapeutic agents for this disorder. RECENT FINDINGS: A growing body of evidence supports that LOAD is a complex disorder, causally associated to a high multiplicity of pathologic mechanisms. New experimental and neuroimaging data, in combination with the recent use of integrative multifactorial data-driven models, support the early role of vascular factors in LOAD genesis and development. Among other relevant roles, the cerebrovascular system has a key modulatory effect on prion-like propagation, deposition and toxicity (e.g. Aß, tau proteins). The early signs of vascular dysregulation during LOAD progression are notable both at the microscopic and the macroscopic scales. SUMMARY: We emphasize that LOAD should be studied as a complex multifactorial disorder, not dominated by a dominant biological factor (e.g. Aß), and without disregarding any relevant pathologic factor, such as vascular dysregulation. Cerebrovascular biomarkers are invaluable for defining multifactorial disease progression models as well as for evaluating the effectiveness of different therapeutic strategies.


Assuntos
Idade de Início , Doença de Alzheimer/etiologia , Transtornos Cerebrovasculares/complicações , Humanos
13.
Hum Brain Mapp ; 38(6): 3126-3140, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28321948

RESUMO

Primary patterns in adult brain connectivity are established during development by coordinated networks of transiently expressed genes; however, neural networks remain malleable throughout life. The present study hypothesizes that structural connectivity from key seed regions may induce effects on their connected targets, which are reflected in gene expression at those targeted regions. To test this hypothesis, analyses were performed on data from two brains from the Allen Human Brain Atlas, for which both gene expression and DW-MRI were available. Structural connectivity was estimated from the DW-MRI data and an approach motivated by network topology, that is, weighted gene coexpression network analysis (WGCNA), was used to cluster genes with similar patterns of expression across the brain. Group exponential lasso models were then used to predict gene cluster expression summaries as a function of seed region structural connectivity patterns. In several gene clusters, brain regions located in the brain stem, diencephalon, and hippocampal formation were identified that have significant predictive power for these expression summaries. These connectivity-associated clusters are enriched in genes associated with synaptic signaling and brain plasticity. Furthermore, using seed region based connectivity provides a novel perspective in understanding relationships between gene expression and connectivity. Hum Brain Mapp 38:3126-3140, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Encéfalo/metabolismo , Expressão Gênica/fisiologia , Redes Reguladoras de Genes/fisiologia , Vias Neurais/metabolismo , Adulto , Encéfalo/citologia , Análise por Conglomerados , Conectoma , Conjuntos de Dados como Assunto , Imagem de Difusão por Ressonância Magnética , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Adulto Jovem
14.
Neurobiol Learn Mem ; 141: 33-43, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28323202

RESUMO

The structural connectome provides relevant information about experience and training-related changes in the brain. Here, we used network-based statistics (NBS) and graph theoretical analyses to study structural changes in the brain as a function of cognitive training. Fifty-six young women were divided in two groups (experimental and control). We assessed their cognitive function before and after completing a working memory intervention using a comprehensive battery that included fluid and crystallized abilities, working memory and attention control, and we also obtained structural MRI images. We acquired and analyzed diffusion-weighted images to reconstruct the anatomical connectome and we computed standardized changes in connectivity as well as group differences across time using NBS. We also compared group differences relying on a variety of graph-theory indices (clustering, characteristic path length, global and local efficiency and strength) for the whole network as well as for the sub-network derived from NBS analyses. Finally, we calculated correlations between these graph indices and training performance as well as the behavioral changes in cognitive function. Our results revealed enhanced connectivity for the training group within one specific network comprised of nodes/regions supporting cognitive processes required by the training (working memory, interference resolution, inhibition, and task engagement). Significant group differences were also observed for strength and global efficiency indices in the sub-network detected by NBS. Therefore, the connectome approach is a valuable method for tracking the effects of cognitive training interventions across specific sub-networks. Moreover, this approach allowsfor the computation of graph theoretical network metricstoquantifythetopological architecture of the brain networkdetected. The observed structural brain changes support the behavioral results reported earlier (see Colom, Román, et al., 2013).


Assuntos
Atenção/fisiologia , Encéfalo/fisiologia , Cognição/fisiologia , Conectoma , Memória de Curto Prazo/fisiologia , Rede Nervosa/fisiologia , Adolescente , Encéfalo/diagnóstico por imagem , Feminino , Humanos , Inibição Psicológica , Imageamento por Ressonância Magnética , Rede Nervosa/diagnóstico por imagem , Testes Neuropsicológicos , Adulto Jovem
15.
Hum Brain Mapp ; 36(2): 577-90, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25324040

RESUMO

Resting-state studies conducted with stroke patients are scarce. The study of brain activity and connectivity at rest provides a unique opportunity for the investigation of brain rewiring after stroke and plasticity changes. This study sought to identify dynamic changes in the functional organization of the default mode network (DMN) of stroke patients at three months after stroke. Eleven patients (eight male and three female; age range: 48-72) with right cortical and subcortical ischemic infarctions and 17 controls (eleven males and six females; age range: 57-69) were assessed by neurological and neuropsychological examinations and scanned with resting-state functional magnetic ressonance imaging. First, we explored group differences in functional activity within the DMN by means of probabilistic independent component analysis followed by a dual regression approach. Second, we estimated functional connectivity between 11 DMN nodes both locally by means of seed-based connectivity analysis, as well as globally by means of graph-computation analysis. We found that patients had greater DMN activity in the left precuneus and the left anterior cingulate gyrus when compared with healthy controls (P < 0.05 family-wise error corrected). Seed-based connectivity analysis showed that stroke patients had significant impairment (P = 0.014; threshold = 2.00) in the connectivity between the following five DMN nodes: left superior frontal gyrus (lSFG) and posterior cingulate cortex (t = 2.01); left parahippocampal gyrus and right superior frontal gyrus (t = 2.11); left parahippocampal gyrus and lSFG (t = 2.39); right parietal and lSFG (t = 2.29). Finally, mean path length obtained from graph-computation analysis showed positive correlations with semantic fluency test (r(s) = 0.454; P = 0.023), phonetic fluency test (r(s) = 0.523; P = 0.007) and the mini mental state examination (r(s) = 0.528; P = 0.007). In conclusion, the ability to regulate activity of the DMN appears to be a central part of normal brain function in stroke patients. Our study expands the understanding of the changes occurring in the brain after stroke providing a new avenue for investigating lesion-induced network plasticity.


Assuntos
Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/psicologia , Encéfalo/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/psicologia , Adulto , Idoso , Mapeamento Encefálico , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Vias Neurais/fisiopatologia , Testes Neuropsicológicos , Descanso , Processamento de Sinais Assistido por Computador
16.
PLoS Comput Biol ; 10(11): e1003956, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25412207

RESUMO

Misfolded proteins (MP) are a key component in aging and associated neurodegenerative disorders. For example, misfolded Amyloid-ß (Aß) and tau proteins are two neuropathogenic hallmarks of Alzheimer's disease. Mechanisms underlying intra-brain MP propagation/deposition remain essentially uncharacterized. Here, is introduced an epidemic spreading model (ESM) for MP dynamics that considers propagation-like interactions between MP agents and the brain's clearance response across the structural connectome. The ESM reproduces advanced Aß deposition patterns in the human brain (explaining 46∼56% of the variance in regional Aß loads, in 733 subjects from the ADNI database). Furthermore, this model strongly supports a) the leading role of Aß clearance deficiency and early Aß onset age during Alzheimer's disease progression, b) that effective anatomical distance from Aß outbreak region explains regional Aß arrival time and Aß deposition likelihood, c) the multi-factorial impact of APOE e4 genotype, gender and educational level on lifetime intra-brain Aß propagation, and d) the modulatory impact of Aß propagation history on tau proteins concentrations, supporting the hypothesis of an interrelated pathway between Aß pathophysiology and tauopathy. To our knowledge, the ESM is the first computational model highlighting the direct link between structural brain networks, production/clearance of pathogenic proteins and associated intercellular transfer mechanisms, individual genetic/demographic properties and clinical states in health and disease. In sum, the proposed ESM constitutes a promising framework to clarify intra-brain region to region transference mechanisms associated with aging and neurodegenerative disorders.


Assuntos
Envelhecimento/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doenças Neurodegenerativas/metabolismo , Adolescente , Adulto , Peptídeos beta-Amiloides/química , Encéfalo/metabolismo , Epidemias , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Príons/química , Príons/metabolismo , Dobramento de Proteína , Adulto Jovem
17.
Neuroimage ; 84: 495-504, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24018306

RESUMO

How the brain deals with more than one language and whether we need different or extra brain language sub-networks to support more than one language remain unanswered questions. Here, we investigate structural brain network differences between early bilinguals and monolinguals. Using diffusion-weighted MRI (DW-MRI) tractography techniques and a network-based statistic (NBS) procedure, we found two structural sub-networks more connected by white matter (WM) tracts in bilinguals than in monolinguals; confirming WM brain plasticity in bilinguals. One of these sub-networks comprises left frontal and parietal/temporal regions, while the other comprises left occipital and parietal/temporal regions and also the right superior frontal gyrus. Most of these regions have been related to language processing and monitoring; suggesting that bilinguals develop specialized language sub-networks to deal with the two languages. Additionally, a complex network analysis showed that these sub-networks are more graph-efficient in bilinguals than monolinguals and this increase seems to be at the expense of a whole-network graph-efficiency decrease.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Conectoma/métodos , Multilinguismo , Rede Nervosa/anatomia & histologia , Rede Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Adulto , Imagem de Tensor de Difusão/métodos , Feminino , Humanos , Masculino , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espanha , Adulto Jovem
18.
Sci Rep ; 14(1): 7269, 2024 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-38538816

RESUMO

Typical differential single-nucleus gene expression (snRNA-seq) analyses in Alzheimer's disease (AD) provide fixed snapshots of cellular alterations, making the accurate detection of temporal cell changes challenging. To characterize the dynamic cellular and transcriptomic differences in AD neuropathology, we apply the novel concept of RNA velocity to the study of single-nucleus RNA from the cortex of 60 subjects with varied levels of AD pathology. RNA velocity captures the rate of change of gene expression by comparing intronic and exonic sequence counts. We performed differential analyses to find the significant genes driving both cell type-specific RNA velocity and expression differences in AD, extensively compared these two transcriptomic metrics, and clarified their associations with multiple neuropathologic traits. The results were cross-validated in an independent dataset. Comparison of AD pathology-associated RNA velocity with parallel gene expression differences reveals sets of genes and molecular pathways that underlie the dynamic and static regimes of cell type-specific dysregulations underlying the disease. Differential RNA velocity and its linked progressive neuropathology point to significant dysregulations in synaptic organization and cell development across cell types. Notably, most of the genes underlying this synaptic dysregulation showed increased RNA velocity in AD subjects compared to controls. Accelerated cell changes were also observed in the AD subjects, suggesting that the precocious depletion of precursor cell pools might be associated with neurodegeneration. Overall, this study uncovers active molecular drivers of the spatiotemporal alterations in AD and offers novel insights towards gene- and cell-centric therapeutic strategies accounting for dynamic cell perturbations and synaptic disruptions.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , RNA/genética , Transcriptoma/genética , Perfilação da Expressão Gênica , Núcleo Solitário/metabolismo
19.
Elife ; 122024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38512130

RESUMO

For over a century, brain research narrative has mainly centered on neuron cells. Accordingly, most neurodegenerative studies focus on neuronal dysfunction and their selective vulnerability, while we lack comprehensive analyses of other major cell types' contribution. By unifying spatial gene expression, structural MRI, and cell deconvolution, here we describe how the human brain distribution of canonical cell types extensively predicts tissue damage in 13 neurodegenerative conditions, including early- and late-onset Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, amyotrophic lateral sclerosis, mutations in presenilin-1, and 3 clinical variants of frontotemporal lobar degeneration (behavioral variant, semantic and non-fluent primary progressive aphasia) along with associated three-repeat and four-repeat tauopathies and TDP43 proteinopathies types A and C. We reconstructed comprehensive whole-brain reference maps of cellular abundance for six major cell types and identified characteristic axes of spatial overlapping with atrophy. Our results support the strong mediating role of non-neuronal cells, primarily microglia and astrocytes, in spatial vulnerability to tissue loss in neurodegeneration, with distinct and shared across-disorder pathomechanisms. These observations provide critical insights into the multicellular pathophysiology underlying spatiotemporal advance in neurodegeneration. Notably, they also emphasize the need to exceed the current neuro-centric view of brain diseases, supporting the imperative for cell-specific therapeutic targets in neurodegeneration.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Humanos , Encéfalo , Neurônios , Mapeamento Encefálico
20.
Commun Biol ; 7(1): 591, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38760483

RESUMO

Late onset Alzheimer's disease (AD) is a progressive neurodegenerative disease, with brain changes beginning years before symptoms surface. AD is characterized by neuronal loss, the classic feature of the disease that underlies brain atrophy. However, GWAS reports and recent single-nucleus RNA sequencing (snRNA-seq) efforts have highlighted that glial cells, particularly microglia, claim a central role in AD pathophysiology. Here, we tailor pattern-learning algorithms to explore distinct gene programs by integrating the entire transcriptome, yielding distributed AD-predictive modules within the brain's major cell-types. We show that these learned modules are biologically meaningful through the identification of new and relevant enriched signaling cascades. The predictive nature of our modules, especially in microglia, allows us to infer each subject's progression along a disease pseudo-trajectory, confirmed by post-mortem pathological brain tissue markers. Additionally, we quantify the interplay between pairs of cell-type modules in the AD brain, and localized known AD risk genes to enriched module gene programs. Our collective findings advocate for a transition from cell-type-specificity to gene modules specificity to unlock the potential of unique gene programs, recasting the roles of recently reported genome-wide AD risk loci.


Assuntos
Doença de Alzheimer , Progressão da Doença , Transcriptoma , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Humanos , Encéfalo/metabolismo , Encéfalo/patologia , Microglia/metabolismo , Microglia/patologia , Perfilação da Expressão Gênica , Redes Reguladoras de Genes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA