Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
bioRxiv ; 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39314329

RESUMO

Amyloid-beta (Aß) plaques and surrounding glial activation are prominent histopathological hallmarks of Alzheimer's Disease (AD). However, it is unclear how Aß plaques interact with surrounding glial cells in the human brain. Here, we applied spatial transcriptomics (ST) and immunohistochemistry (IHC) for Aß, GFAP, and IBA1 to acquire data from 258,987 ST spots within 78 postmortem brain sections of 21 individuals. By coupling ST and adjacent-section IHC, we showed that low Aß spots exhibit transcriptomic profiles indicative of greater neuronal loss than high Aß spots, and high-glia spots present transcriptomic changes indicative of more significant inflammation and neurodegeneration. Furthermore, we observed that this ST glial response bears signatures of reported mouse gene modules of plaque-induced genes (PIG), oligodendrocyte (OLIG) response, disease-associated microglia (DAM), and disease-associated astrocytes (DAA), as well as different microglia (MG) states identified in human AD brains, indicating that multiple glial cell states arise around plaques and contribute to local immune response. We then validated the observed effects of Aß on cell apoptosis and plaque-surrounding glia on inflammation and synaptic loss using IHC. In addition, transcriptomic changes of iPSC-derived microglia-like cells upon short-interval Aß treatment mimic the ST glial response and mirror the reported activated MG states. Our results demonstrate an exacerbation of synaptic and neuronal loss in low-Aß or high-glia areas, indicating that microglia response to Aß-oligomers likely initiates glial activation in plaque-glia niches. Our study lays the groundwork for future pathology genomics studies, opening the door for investigating pathological heterogeneity and causal effects in neurodegenerative diseases.

2.
medRxiv ; 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39185527

RESUMO

Advances have led to a greater understanding of the genetics of Alzheimer's Disease (AD). However, the gap between the predicted and observed genetic heritability estimates when using single nucleotide polymorphisms (SNPs) and small indel data remains. Large genomic rearrangements, known as structural variants (SVs), have the potential to account for this missing genetic heritability. By leveraging data from two ongoing cohort studies of aging and dementia, the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP), we performed genome-wide association analysis testing around 20,000 common SVs from 1,088 participants with whole genome sequencing (WGS) data. A range of Alzheimer's Disease and Related Disorders (AD/ADRD) clinical and pathologic traits were examined. Given the limited sample size, no genome-wide significant association was found, but we mapped SVs across 81 AD risk loci and discovered 22 SVs in linkage disequilibrium (LD) with GWAS lead variants and directly associated with AD/ADRD phenotypes (nominal P < 0.05). The strongest association was a deletion of an Alu element in the 3'UTR of the TMEM106B gene. This SV was in high LD with the respective AD GWAS locus and was associated with multiple AD/ADRD phenotypes, including tangle density, TDP-43, and cognitive resilience. The deletion of this element was also linked to lower TMEM106B protein abundance. We also found a 22 kb deletion associated with depression in ROSMAP and bearing similar association patterns as AD GWAS SNPs at the IQCK locus. In addition, genome-wide scans allowed the identification of 7 SVs, with no LD with SNPs and nominally associated with AD/ADRD traits. This result suggests potentially new ADRD risk loci not discoverable using SNP data. Among these findings, we highlight a 5.6 kb duplication of coding regions of the gene C1orf186 at chromosome 1 associated with indices of cognitive impairment, decline, and resilience. While further replication in independent datasets is needed to validate these findings, our results support the potential roles of common structural variations in the pathogenesis of AD/ADRD.

3.
Nat Neurosci ; 2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39187705

RESUMO

Proteomics can shed light on the dynamic and multifaceted alterations in neurodegenerative disorders like Alzheimer's disease (AD). Combining radioligands measuring ß-amyloid (Aß) plaques and tau tangles with cerebrospinal fluid proteomics, we uncover molecular events mirroring different stages of AD pathology in living humans. We found 127 differentially abundant proteins (DAPs) across the AD spectrum. The strongest Aß-related proteins were mainly expressed in glial cells and included SMOC1 and ITGAM. A dozen proteins linked to ATP metabolism and preferentially expressed in neurons were independently associated with tau tangle load and tau accumulation. Only 20% of the DAPs were also altered in other neurodegenerative diseases, underscoring AD's distinct proteome. Two co-expression modules related, respectively, to protein metabolism and microglial immune response encompassed most DAPs, with opposing, staggered trajectories along the AD continuum. We unveil protein signatures associated with Aß and tau proteinopathy in vivo, offering insights into complex neural responses and potential biomarkers and therapeutics targeting different disease stages.

4.
medRxiv ; 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38947084

RESUMO

The pathophysiology underlying various manifestations of cerebral small vessel disease (cSVD) remains obscure. Using cerebrospinal fluid proximity extension assays and co-expression network analysis of 2,943 proteins, we found common and distinct proteomic signatures between white matter lesions (WML), microbleeds and infarcts measured in 856 living patients, and validated WML-associated proteins in three additional datasets. Proteins indicative of extracellular matrix dysregulation and vascular remodeling, including ELN, POSTN, CCN2 and MMP12 were elevated across all cSVD manifestations, with MMP12 emerging as an early cSVD indicator. cSVD-associated proteins formed a co-abundance network linked to metabolism and enriched in endothelial and arterial smooth muscle cells, showing elevated levels at early disease manifestations. Later disease stages involved changes in microglial proteins, associated with longitudinal WML progression, and changes in neuronal proteins mediating WML-associated cognitive decline. These findings provide an atlas of novel cSVD biomarkers and a promising roadmap for the next generation of cSVD therapeutics.

5.
Nat Commun ; 14(1): 7036, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37923721

RESUMO

Emerging evidence shows that the meninges conduct essential immune surveillance and immune defense at the brain border, and the dysfunction of meningeal immunity contributes to aging and neurodegeneration. However, no study exists on the molecular properties of cell types within human leptomeninges. Here, we provide single nuclei profiling of dissected postmortem leptomeninges from aged individuals. We detect diverse cell types, including unique meningeal endothelial, mural, and fibroblast subtypes. For immune cells, we show that most T cells express CD8 and bear characteristics of tissue-resident memory T cells. We also identify distinct subtypes of border-associated macrophages (BAMs) that display differential gene expressions from microglia and express risk genes for Alzheimer's Disease (AD), as nominated by genome-wide association studies (GWAS). We discover cell-type-specific differentially expressed genes in individuals with Alzheimer's dementia, particularly in fibroblasts and BAMs. Indeed, when cultured, leptomeningeal cells display the signature of ex vivo AD fibroblasts upon amyloid-ß treatment. We further explore ligand-receptor interactions within the leptomeningeal niche and computationally infer intercellular communications in AD. Thus, our study establishes a molecular map of human leptomeningeal cell types, providing significant insight into the border immune and fibrotic responses in AD.


Assuntos
Doença de Alzheimer , Estudo de Associação Genômica Ampla , Humanos , Idoso , Meninges , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Macrófagos/metabolismo , Envelhecimento , Microglia/metabolismo
6.
Cell Rep ; 42(11): 113439, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37963017

RESUMO

Human brain size changes dynamically through early development, peaks in adolescence, and varies up to 2-fold among adults. However, the molecular genetic underpinnings of interindividual variation in brain size remain unknown. Here, we leveraged postmortem brain RNA sequencing and measurements of brain weight (BW) in 2,531 individuals across three independent datasets to identify 928 genome-wide significant associations with BW. Genes associated with higher or lower BW showed distinct neurodevelopmental trajectories and spatial patterns that mapped onto functional and cellular axes of brain organization. Expression of BW genes was predictive of interspecies differences in brain size, and bioinformatic annotation revealed enrichment for neurogenesis and cell-cell communication. Genome-wide, transcriptome-wide, and phenome-wide association analyses linked BW gene sets to neuroimaging measurements of brain size and brain-related clinical traits. Cumulatively, these results represent a major step toward delineating the molecular pathways underlying human brain size variation in health and disease.


Assuntos
Encéfalo , Transcriptoma , Adulto , Humanos , Tamanho do Órgão , Encéfalo/metabolismo , Fenótipo , Estudo de Associação Genômica Ampla/métodos , Biologia Molecular , Predisposição Genética para Doença
7.
Genome Biol ; 24(1): 228, 2023 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828545

RESUMO

Clustering molecular data into informative groups is a primary step in extracting robust conclusions from big data. However, due to foundational issues in how they are defined and detected, such clusters are not always reliable, leading to unstable conclusions. We compare popular clustering algorithms across thousands of synthetic and real biological datasets, including a new consensus clustering algorithm-SpeakEasy2: Champagne. These tests identify trends in performance, show no single method is universally optimal, and allow us to examine factors behind variation in performance. Multiple metrics indicate SpeakEasy2 generally provides robust, scalable, and informative clusters for a range of applications.


Assuntos
Algoritmos , Perfilação da Expressão Gênica , Perfilação da Expressão Gênica/métodos , Análise por Conglomerados , Big Data
8.
Entropy (Basel) ; 25(8)2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37628148

RESUMO

Mapping network nodes and edges to communities and network functions is crucial to gaining a higher level of understanding of the network structure and functions. Such mappings are particularly challenging to design for covert social networks, which intentionally hide their structure and functions to protect important members from attacks or arrests. Here, we focus on correctly inferring the structures and functions of such networks, but our methodology can be broadly applied. Without the ground truth, knowledge about the allocation of nodes to communities and network functions, no single network based on the noisy data can represent all plausible communities and functions of the true underlying network. To address this limitation, we apply a generative model that randomly distorts the original network based on the noisy data, generating a pool of statistically equivalent networks. Each unique generated network is recorded, while each duplicate of the already recorded network just increases the repetition count of that network. We treat each such network as a variant of the ground truth with the probability of arising in the real world approximated by the ratio of the count of this network's duplicates plus one to the total number of all generated networks. Communities of variants with frequently occurring duplicates contain persistent patterns shared by their structures. Using Shannon entropy, we can find a variant that minimizes the uncertainty for operations planned on the network. Repeatedly generating new pools of networks from the best network of the previous step for several steps lowers the entropy of the best new variant. If the entropy is too high, the network operators can identify nodes, the monitoring of which can achieve the most significant reduction in entropy. Finally, we also present a heuristic for constructing a new variant, which is not randomly generated but has the lowest expected cost of operating on the distorted mappings of network nodes to communities and functions caused by noisy data.

9.
bioRxiv ; 2023 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-37546752

RESUMO

Neuroimaging is commonly used to infer human brain connectivity, but those measurements are far-removed from the molecular underpinnings at synapses. To uncover the molecular basis of human brain connectivity, we analyzed a unique cohort of 98 individuals who provided neuroimaging and genetic data contemporaneous with dendritic spine morphometric, proteomic, and gene expression data from the superior frontal and inferior temporal gyri. Through cellular contextualization of the molecular data with dendritic spine morphology, we identified hundreds of proteins related to synapses, energy metabolism, and RNA processing that explain between-individual differences in functional connectivity and structural covariation. By integrating data at the genetic, molecular, subcellular, and tissue levels, we bridged the divergent fields of molecular biology and neuroimaging to identify a molecular basis of brain connectivity. One-Sentence Summary: Dendritic spine morphometry and synaptic proteins unite the divergent fields of molecular biology and neuroimaging.

10.
Nat Commun ; 13(1): 655, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115553

RESUMO

Identifying the molecular systems and proteins that modify the progression of Alzheimer's disease and related dementias (ADRD) is central to drug target selection. However, discordance between mRNA and protein abundance, and the scarcity of proteomic data, has limited our ability to advance candidate targets that are mainly based on gene expression. Therefore, by using a deep neural network that predicts protein abundance from mRNA expression, here we attempt to track the early protein drivers of ADRD. Specifically, by applying the clei2block deep learning model to 1192 brain RNA-seq samples, we identify protein modules and disease-associated expression changes that were not directly observed at the mRNA level. Moreover, pseudo-temporal trajectory inference based on the predicted proteome became more closely correlated with cognitive decline and hippocampal atrophy compared to RNA-based trajectories. This suggests that the predicted changes in protein expression could provide a better molecular representation of ADRD progression. Furthermore, overlaying clinical traits on protein pseudotime trajectory identifies protein modules altered before cognitive impairment. These results demonstrate how our method can be used to identify potential early protein drivers and possible drug targets for treating and/or preventing ADRD.


Assuntos
Doença de Alzheimer/genética , Demência/genética , Redes Neurais de Computação , Proteoma/genética , Proteômica/métodos , RNA Mensageiro/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Aprendizado Profundo , Demência/metabolismo , Feminino , Humanos , Masculino , Espectrometria de Massas/métodos , Biossíntese de Proteínas , Proteoma/metabolismo , RNA Mensageiro/metabolismo , RNA-Seq/métodos , Transcriptoma/genética
11.
Nat Neurosci ; 25(2): 213-225, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35115731

RESUMO

The biological processes that are disrupted in the Alzheimer's disease (AD) brain remain incompletely understood. In this study, we analyzed the proteomes of more than 1,000 brain tissues to reveal new AD-related protein co-expression modules that were highly preserved across cohorts and brain regions. Nearly half of the protein co-expression modules, including modules significantly altered in AD, were not observed in RNA networks from the same cohorts and brain regions, highlighting the proteopathic nature of AD. Two such AD-associated modules unique to the proteomic network included a module related to MAPK signaling and metabolism and a module related to the matrisome. The matrisome module was influenced by the APOE ε4 allele but was not related to the rate of cognitive decline after adjustment for neuropathology. By contrast, the MAPK/metabolism module was strongly associated with the rate of cognitive decline. Disease-associated modules unique to the proteome are sources of promising therapeutic targets and biomarkers for AD.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Disfunção Cognitiva/patologia , Humanos , Proteoma , Proteômica , RNA/metabolismo
12.
Neuron ; 109(21): 3402-3420.e9, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34473944

RESUMO

We have generated a controlled and manipulable resource that captures genetic risk for Alzheimer's disease: iPSC lines from 53 individuals coupled with RNA and proteomic profiling of both iPSC-derived neurons and brain tissue of the same individuals. Data collected for each person include genome sequencing, longitudinal cognitive scores, and quantitative neuropathology. The utility of this resource is exemplified here by analyses of neurons derived from these lines, revealing significant associations between specific Aß and tau species and the levels of plaque and tangle deposition in the brain and, more importantly, with the trajectory of cognitive decline. Proteins and networks are identified that are associated with AD phenotypes in iPSC neurons, and relevant associations are validated in brain. The data presented establish this iPSC collection as a resource for investigating person-specific processes in the brain that can aid in identifying and validating molecular pathways underlying AD.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Idoso , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Cognição , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Proteômica , Proteínas tau/genética , Proteínas tau/metabolismo
13.
Sci Rep ; 11(1): 11311, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050212

RESUMO

Motor resilience proteins may be a high value therapeutic target that offset the negative effects of pathologies on motor function. This study sought to identify cortical proteins associated with motor decline unexplained by brain pathologies that provide motor resilience. We studied 1226 older decedents with annual motor testing, postmortem brain pathologies and quantified 226 proteotypic peptides in prefrontal cortex. Twenty peptides remained associated with motor decline in models controlling for ten brain pathologies (FDR < 0.05). Higher levels of nine peptides and lower levels of eleven peptides were related to slower decline. A higher motor resilience protein score based on averaging the levels of all 20 peptides was related to slower motor decline, less severe parkinsonism and lower odds of mobility disability before death. Cortical proteins may provide motor resilience. Targeting these proteins in further drug discovery may yield novel interventions to maintain motor function in old age.


Assuntos
Transtornos dos Movimentos/metabolismo , Peptídeos/metabolismo , Córtex Pré-Frontal/metabolismo , Desempenho Psicomotor , Feminino , Humanos , Masculino , Transtornos dos Movimentos/etiologia , Córtex Pré-Frontal/patologia , Estudos Prospectivos
14.
Transl Psychiatry ; 11(1): 50, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446646

RESUMO

Microglial dysfunction has been proposed as one of the many cellular mechanisms that can contribute to the development of Alzheimer's disease (AD). Here, using a transcriptional network map of the human frontal cortex, we identify five modules of co-expressed genes related to microglia and assess their role in the neuropathologic features of AD in 540 subjects from two cohort studies of brain aging. Two of these transcriptional programs-modules 113 and 114-relate to the accumulation of ß-amyloid, while module 5 relates to tau pathology. We replicate these associations in brain epigenomic data and in two independent datasets. In terms of tau, we propose that module 5, a marker of activated microglia, may lead to tau accumulation and subsequent cognitive decline. We validate our model further by showing that three representative module 5 genes (ACADVL, TRABD, and VASP) encode proteins that are upregulated in activated microglia in AD.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Humanos , Microglia/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Learn Mem ; 27(9): 355-371, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32817302

RESUMO

Individual differences in cognitive decline during normal aging and Alzheimer's disease (AD) are common, but the molecular mechanisms underlying these distinct outcomes are not fully understood. We utilized a combination of genetic, molecular, and behavioral data from a mouse population designed to model human variation in cognitive outcomes to search for the molecular mechanisms behind this population-wide variation. Specifically, we used a systems genetics approach to relate gene expression to cognitive outcomes during AD and normal aging. Statistical causal-inference Bayesian modeling was used to model systematic genetic perturbations matched with cognitive data that identified astrocyte and microglia molecular networks as drivers of cognitive resilience to AD. Using genetic mapping, we identified Fgf2 as a potential regulator of the astrocyte network associated with individual differences in short-term memory. We also identified several immune genes as regulators of a microglia network associated with individual differences in long-term memory, which was partly mediated by amyloid burden. Finally, significant overlap between mouse and two different human coexpression networks provided strong evidence of translational relevance for the genetically diverse AD-BXD panel as a model of late-onset AD. Together, this work identified two candidate molecular pathways enriched for microglia and astrocyte genes that serve as causal AD cognitive biomarkers, and provided a greater understanding of processes that modulate individual and population-wide differences in cognitive outcomes during AD.


Assuntos
Envelhecimento , Doença de Alzheimer , Astrócitos , Disfunção Cognitiva , Reserva Cognitiva , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Microglia , Envelhecimento/genética , Envelhecimento/imunologia , Envelhecimento/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Comportamento Animal/fisiologia , Biomarcadores , Encéfalo , Disfunção Cognitiva/genética , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Reserva Cognitiva/fisiologia , Feminino , Humanos , Individualidade , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos
16.
PLoS Comput Biol ; 16(8): e1008120, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32804935

RESUMO

Complexity of cell-type composition has created much skepticism surrounding the interpretation of bulk tissue transcriptomic studies. Recent studies have shown that deconvolution algorithms can be applied to computationally estimate cell-type proportions from gene expression data of bulk blood samples, but their performance when applied to brain tissue is unclear. Here, we have generated an immunohistochemistry (IHC) dataset for five major cell-types from brain tissue of 70 individuals, who also have bulk cortical gene expression data. With the IHC data as the benchmark, this resource enables quantitative assessment of deconvolution algorithms for brain tissue. We apply existing deconvolution algorithms to brain tissue by using marker sets derived from human brain single cell and cell-sorted RNA-seq data. We show that these algorithms can indeed produce informative estimates of constituent cell-type proportions. In fact, neuronal subpopulations can also be estimated from bulk brain tissue samples. Further, we show that including the cell-type proportion estimates as confounding factors is important for reducing false associations between Alzheimer's disease phenotypes and gene expression. Lastly, we demonstrate that using more accurate marker sets can substantially improve statistical power in detecting cell-type specific expression quantitative trait loci (eQTLs).


Assuntos
Algoritmos , Encéfalo , Perfilação da Expressão Gênica/métodos , Análise de Sequência de RNA/métodos , Transcriptoma/genética , Encéfalo/citologia , Encéfalo/metabolismo , Biologia Computacional , Humanos , Imuno-Histoquímica , Especificidade de Órgãos/genética , Fenótipo , Locos de Características Quantitativas/genética , Análise de Célula Única
17.
Nat Mach Intell ; 2(7): 376-386, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32671330

RESUMO

Identifying the molecular mechanisms that control differential gene expression (DE) is a major goal of basic and disease biology. We develop a systems biology model to predict DE, and mine the biological basis of the factors that influence predicted gene expression, in order to understand how it may be generated. This model, called DEcode, utilizes deep learning to predict DE based on genome-wide binding sites on RNAs and promoters. Ranking predictive factors from the DEcode indicates that clinically relevant expression changes between thousands of individuals can be predicted mainly through the joint action of post-transcriptional RNA-binding factors. We also show the broad potential applications of DEcode to generate biological insights, by predicting DE between tissues, differential transcript-usage, and drivers of aging throughout the human lifespan, of gene coexpression relationships on a genome-wide scale, and of frequently DE genes across diverse conditions. Researchers can freely utilize DEcode to identify influential molecular mechanisms for any human expression data - www.differentialexpression.org.

18.
Cell Rep ; 32(2): 107908, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32668255

RESUMO

We present a consensus atlas of the human brain transcriptome in Alzheimer's disease (AD), based on meta-analysis of differential gene expression in 2,114 postmortem samples. We discover 30 brain coexpression modules from seven regions as the major source of AD transcriptional perturbations. We next examine overlap with 251 brain differentially expressed gene sets from mouse models of AD and other neurodegenerative disorders. Human-mouse overlaps highlight responses to amyloid versus tau pathology and reveal age- and sex-dependent expression signatures for disease progression. Human coexpression modules enriched for neuronal and/or microglial genes broadly overlap with mouse models of AD, Huntington's disease, amyotrophic lateral sclerosis, and aging. Other human coexpression modules, including those implicated in proteostasis, are not activated in AD models but rather following other, unexpected genetic manipulations. Our results comprise a cross-species resource, highlighting transcriptional networks altered by human brain pathophysiology and identifying correspondences with mouse models for AD preclinical studies.


Assuntos
Doença de Alzheimer/genética , Encéfalo/metabolismo , Encéfalo/patologia , Transcriptoma/genética , Animais , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , Camundongos , Caracteres Sexuais , Especificidade da Espécie , Transcrição Gênica
19.
Artigo em Inglês | MEDLINE | ID: mdl-31674729

RESUMO

The substantial progress made in the basic sciences of the brain has yet to be adequately translated to successful clinical therapeutics to treat central nervous system (CNS) diseases. Possible explanations include the lack of quantitative and validated biomarkers, the subjective nature of many clinical endpoints, and complex pharmacokinetic/pharmacodynamic relationships, but also the possibility that highly selective drugs in the CNS do not reflect the complex interactions of different brain circuits. Although computational systems pharmacology modeling designed to capture essential components of complex biological systems has been increasingly accepted in pharmaceutical research and development for oncology, inflammation, and metabolic disorders, the uptake in the CNS field has been very modest. In this article, a cross-disciplinary group with representatives from academia, pharma, regulatory, and funding agencies make the case that the identification and exploitation of CNS therapeutic targets for drug discovery and development can benefit greatly from a system and network approach that can span the gap between molecular pathways and the neuronal circuits that ultimately regulate brain activity and behavior. The National Institute of Neurological Disorders and Stroke (NINDS), in collaboration with the National Institute on Aging (NIA), National Institute of Mental Health (NIMH), National Institute on Drug Abuse (NIDA), and National Center for Advancing Translational Sciences (NCATS), convened a workshop to explore and evaluate the potential of a quantitative systems pharmacology (QSP) approach to CNS drug discovery and development. The objective of the workshop was to identify the challenges and opportunities of QSP as an approach to accelerate drug discovery and development in the field of CNS disorders. In particular, the workshop examined the potential for computational neuroscience to perform QSP-based interrogation of the mechanism of action for CNS diseases, along with a more accurate and comprehensive method for evaluating drug effects and optimizing the design of clinical trials. Following up on an earlier white paper on the use of QSP in general disease mechanism of action and drug discovery, this report focuses on new applications, opportunities, and the accompanying limitations of QSP as an approach to drug development in the CNS therapeutic area based on the discussions in the workshop with various stakeholders.


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Doenças do Sistema Nervoso Central/tratamento farmacológico , Desenvolvimento de Medicamentos/métodos , Descoberta de Drogas/métodos , Animais , Humanos , Farmacologia/métodos , Biologia de Sistemas
20.
Transl Psychiatry ; 9(1): 241, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31582723

RESUMO

Alzheimer's disease manifests with both cognitive and motor deficits. However, the degree to which genetic risk of Alzheimer's dementia contributes to late-life motor impairment, and the specific molecular systems underlying these associations, are uncertain. Here, we adopted an integrative multi-omic approach to assess genetic influence on motor impairment in older adults and identified key molecular pathways that may mediate this risk. We built a polygenic risk score for clinical diagnosis of Alzheimer's dementia (AD-PRS) and examined its relationship to several motor phenotypes in 1885 older individuals from two longitudinal aging cohorts. We found that AD-PRS was associated with a previously validated composite motor scores and their components. The major genetic risk factor for sporadic Alzheimer's dementia, the APOE/TOMM40 locus, was not a major driver of these associations. To identify specific molecular features that potentially medicate the genetic risk into motor dysfunction, we examined brain multi-omics, including transcriptome, DNA methylation, histone acetylation (H3K9AC), and targeted proteomics, as well as diverse neuropathologies. We found that a small number of factors account for the majority of the influence of AD-PRS on motor function, which comprises paired helical filament tau-tangle density, H3K9AC in specific chromosomal regions encoding genes involved in neuromuscular process. These multi-omic factors have the potential to elucidate key molecular mechanisms developing motor impairment in the context of Alzheimer's dementia.


Assuntos
Doença de Alzheimer/genética , Encéfalo/patologia , Disfunção Cognitiva/genética , Histonas/genética , Herança Multifatorial , Acetilação , Idoso , Chicago , Estudo de Associação Genômica Ampla , Humanos , Estudos Longitudinais , Testes Neuropsicológicos , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA