Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Neurosci ; 44(29)2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38830764

RESUMO

Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-ß (Aß) pathology (PS2APP) or combined Aß and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.


Assuntos
Doença de Alzheimer , Glicoproteínas de Membrana , Microglia , Esclerose Múltipla , Receptores Imunológicos , Animais , Receptores Imunológicos/agonistas , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Glicoproteínas de Membrana/agonistas , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Camundongos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Feminino , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Anticorpos/farmacologia , Humanos , Peptídeos beta-Amiloides/metabolismo , Proteínas tau/metabolismo
2.
Cell Rep ; 37(13): 110158, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34965428

RESUMO

Non-neuronal responses in neurodegenerative disease have received increasing attention as important contributors to disease pathogenesis and progression. Here we utilize single-cell RNA sequencing to broadly profile 13 cell types in three different mouse models of Alzheimer disease (AD), capturing the effects of tau-only, amyloid-only, or combined tau-amyloid pathology. We highlight microglia, oligodendrocyte, astrocyte, and T cell responses and compare them across these models. Notably, we identify two distinct transcriptional states for oligodendrocytes emerging differentially across disease models, and we determine their spatial distribution. Furthermore, we explore the impact of Trem2 deletion in the context of combined pathology. Trem2 knockout mice exhibit severely blunted microglial responses to combined tau and amyloid pathology, but responses from non-microglial cell types (oligodendrocytes, astrocytes, and T cells) are relatively unchanged. These results delineate core transcriptional states that are engaged in response to AD pathology, and how they are influenced by a key AD risk gene, Trem2.


Assuntos
Doença de Alzheimer/patologia , Amiloide/química , Astrócitos/patologia , Glicoproteínas de Membrana/fisiologia , Oligodendroglia/patologia , Receptores Imunológicos/fisiologia , Linfócitos T/imunologia , Proteínas tau/metabolismo , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodendroglia/imunologia , Oligodendroglia/metabolismo
3.
Neuron ; 109(8): 1283-1301.e6, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33675684

RESUMO

Loss-of-function TREM2 mutations strongly increase Alzheimer's disease (AD) risk. Trem2 deletion has revealed protective Trem2 functions in preclinical models of ß-amyloidosis, a prominent feature of pre-diagnosis AD stages. How TREM2 influences later AD stages characterized by tau-mediated neurodegeneration is unclear. To understand Trem2 function in the context of both ß-amyloid and tau pathologies, we examined Trem2 deficiency in the pR5-183 mouse model expressing mutant tau alone or in TauPS2APP mice, in which ß-amyloid pathology exacerbates tau pathology and neurodegeneration. Single-cell RNA sequencing in these models revealed robust disease-associated microglia (DAM) activation in TauPS2APP mice that was amyloid-dependent and Trem2-dependent. In the presence of ß-amyloid pathology, Trem2 deletion further exacerbated tau accumulation and spreading and promoted brain atrophy. Without ß-amyloid pathology, Trem2 deletion did not affect these processes. Therefore, TREM2 may slow AD progression and reduce tau-driven neurodegeneration by restricting the degree to which ß-amyloid facilitates the spreading of pathogenic tau.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Atrofia/genética , Atrofia/metabolismo , Atrofia/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Receptores Imunológicos/genética , Proteínas tau/genética
4.
Cell Rep ; 31(13): 107843, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32610143

RESUMO

Damage-associated microglia (DAM) profiles observed in Alzheimer's disease (AD)-related mouse models reflect an activation state that could modulate AD risk or progression. To learn whether human AD microglia (HAM) display a similar profile, we develop a method for purifying cell types from frozen cerebrocortical tissues for RNA-seq analysis, allowing better transcriptome coverage than typical single-nucleus RNA-seq approaches. The HAM profile we observe bears little resemblance to the DAM profile. Instead, HAM display an enhanced human aging profile, in addition to other disease-related changes such as APOE upregulation. Analyses of whole-tissue RNA-seq and single-cell/nucleus RNA-seq datasets corroborate our findings and suggest that the lack of DAM response in human microglia occurs specifically in AD tissues, not other neurodegenerative settings. These results, which can be browsed at http://research-pub.gene.com/BrainMyeloidLandscape, provide a genome-wide picture of microglial activation in human AD and highlight considerable differences between mouse models and human disease.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Senescência Celular/genética , Microglia/metabolismo , Microglia/patologia , Ativação Transcricional/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Bases de Dados Genéticas , Feminino , Lobo Frontal/patologia , Secções Congeladas , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Xenoenxertos , Humanos , Masculino , Camundongos , Monócitos/metabolismo , Esclerose Múltipla/patologia , Fenótipo , Reprodutibilidade dos Testes , Fatores de Risco , Lobo Temporal/patologia
5.
J Neurosci ; 40(9): 1956-1974, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-31980586

RESUMO

TREM2 is an Alzheimer's disease (AD) risk gene expressed in microglia. To study the role of Trem2 in a mouse model of ß-amyloidosis, we compared PS2APP transgenic mice versus PS2APP mice lacking Trem2 (PS2APP;Trem2ko) at ages ranging from 4 to 22 months. Microgliosis was impaired in PS2APP;Trem2ko mice, with Trem2-deficient microglia showing compromised expression of proliferation/Wnt-related genes and marked accumulation of ApoE. Plaque abundance was elevated in PS2APP;Trem2ko females at 6-7 months; but by 12 or 19-22 months of age, it was notably diminished in female and male PS2APP;Trem2ko mice, respectively. Across all ages, plaque morphology was more diffuse in PS2APP;Trem2ko brains, and the Aß42:Aß40 ratio was elevated. The amount of soluble, fibrillar Aß oligomers also increased in PS2APP;Trem2ko hippocampi. Associated with these changes, axonal dystrophy was exacerbated from 6 to 7 months onward in PS2APP;Trem2ko mice, notwithstanding the reduced plaque load at later ages. PS2APP;Trem2ko mice also exhibited more dendritic spine loss around plaque and more neurofilament light chain in CSF. Thus, aggravated neuritic dystrophy is a more consistent outcome of Trem2 deficiency than amyloid plaque load, suggesting that the microglial packing of Aß into dense plaque is an important neuroprotective activity.SIGNIFICANCE STATEMENT Genetic studies indicate that TREM2 gene mutations confer increased Alzheimer's disease (AD) risk. We studied the effects of Trem2 deletion in the PS2APP mouse AD model, in which overproduction of Aß peptide leads to amyloid plaque formation and associated neuritic dystrophy. Interestingly, neuritic dystrophies were intensified in the brains of Trem2-deficient mice, despite these mice displaying reduced plaque accumulation at later ages (12-22 months). Microglial clustering around plaques was impaired, plaques were more diffuse, and the Aß42:Aß40 ratio and amount of soluble, fibrillar Aß oligomers were elevated in Trem2-deficient brains. These results suggest that the Trem2-dependent compaction of Aß into dense plaques is a protective microglial activity, limiting the exposure of neurons to toxic Aß species.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Axônios/patologia , Espinhas Dendríticas/patologia , Glicoproteínas de Membrana/genética , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/genética , Receptores Imunológicos/genética , Fator Trefoil-1/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Neuritos/patologia , Proteínas de Neurofilamentos/líquido cefalorraquidiano , Placa Amiloide/patologia
6.
J Neurosci ; 40(5): 958-973, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31831521

RESUMO

Cortical circuit activity is shaped by the parvalbumin (PV) and somatostatin (SST) interneurons that inhibit principal excitatory (EXC) neurons and the vasoactive intestinal peptide (VIP) interneurons that suppress activation of other interneurons. To understand the molecular-genetic basis of functional specialization and identify potential drug targets specific to each neuron subtype, we performed a genome wide assessment of both gene expression and splicing across EXC, PV, SST and VIP neurons from male and female mouse brains. These results reveal numerous examples where neuron subtype-specific gene expression, as well as splice-isoform usage, can explain functional differences between neuron subtypes, including in presynaptic plasticity, postsynaptic receptor function, and synaptic connectivity specification. We provide a searchable web resource for exploring differential mRNA expression and splice form usage between excitatory, PV, SST, and VIP neurons (http://research-pub.gene.com/NeuronSubtypeTranscriptomes). This resource, combining a unique new dataset and novel application of analysis methods to multiple relevant datasets, identifies numerous potential drug targets for manipulating circuit function, reveals neuron subtype-specific roles for disease-linked genes, and is useful for understanding gene expression changes observed in human patient brains.SIGNIFICANCE STATEMENT Understanding the basis of functional specialization of neuron subtypes and identifying drug targets for manipulating circuit function requires comprehensive information on cell-type-specific transcriptional profiles. We sorted excitatory neurons and key inhibitory neuron subtypes from mouse brains and assessed differential mRNA expression. We used a genome-wide analysis which not only examined differential gene expression levels but could also detect differences in splice isoform usage. This analysis reveals numerous examples of neuron subtype-specific isoform usage with functional importance, identifies potential drug targets, and provides insight into the neuron subtypes involved in psychiatric disease. We also apply our analysis to two other relevant datasets for comparison, and provide a searchable website for convenient access to the resource.


Assuntos
Córtex Cerebral/metabolismo , Interneurônios/metabolismo , Neurônios/metabolismo , Transcriptoma , Animais , Células Cultivadas , Feminino , Hipocampo/metabolismo , Masculino , Camundongos Transgênicos , Parvalbuminas/metabolismo , RNA Mensageiro/metabolismo , Somatostatina/metabolismo , Peptídeo Intestinal Vasoativo/metabolismo
7.
Cell Rep ; 28(8): 2111-2123.e6, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31433986

RESUMO

Complement pathway overactivation can lead to neuronal damage in various neurological diseases. Although Alzheimer's disease (AD) is characterized by ß-amyloid plaques and tau tangles, previous work examining complement has largely focused on amyloidosis models. We find that glial cells show increased expression of classical complement components and the central component C3 in mouse models of amyloidosis (PS2APP) and more extensively tauopathy (TauP301S). Blocking complement function by deleting C3 rescues plaque-associated synapse loss in PS2APP mice and ameliorates neuron loss and brain atrophy in TauP301S mice, improving neurophysiological and behavioral measurements. In addition, C3 protein is elevated in AD patient brains, including at synapses, and levels and processing of C3 are increased in AD patient CSF and correlate with tau. These results demonstrate that complement activation contributes to neurodegeneration caused by tau pathology and suggest that blocking C3 function might be protective in AD and other tauopathies.


Assuntos
Doença de Alzheimer/imunologia , Amiloidose/imunologia , Complemento C3/metabolismo , Degeneração Neural/imunologia , Tauopatias/imunologia , Doença de Alzheimer/genética , Animais , Atrofia , Comportamento Animal , Biomarcadores/metabolismo , Encéfalo/patologia , Complemento C1q/metabolismo , Complemento C3/líquido cefalorraquidiano , Complemento C3/genética , Modelos Animais de Doenças , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos Transgênicos , Degeneração Neural/genética , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/metabolismo , Sinapses/metabolismo
8.
PLoS Genet ; 14(11): e1007427, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30388101

RESUMO

Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA) is a cell surface inhibitory receptor that recognizes specific O-glycosylated proteins and is expressed on various innate immune cell types including microglia. We show here that a common missense variant (G78R, rs1859788) of PILRA is the likely causal allele for the confirmed Alzheimer's disease risk locus at 7q21 (rs1476679). The G78R variant alters the interaction of residues essential for sialic acid engagement, resulting in >50% reduced binding for several PILRA ligands including a novel ligand, complement component 4A, and herpes simplex virus 1 (HSV-1) glycoprotein B. PILRA is an entry receptor for HSV-1 via glycoprotein B, and macrophages derived from R78 homozygous donors showed significantly decreased levels of HSV-1 infection at several multiplicities of infection compared to homozygous G78 macrophages. We propose that PILRA G78R protects individuals from Alzheimer's disease risk via reduced inhibitory signaling in microglia and reduced microglial infection during HSV-1 recurrence.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Variação Genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Substituição de Aminoácidos , Animais , Loci Gênicos , Humanos , Ligantes , Glicoproteínas de Membrana/química , Camundongos , Modelos Biológicos , Conformação Molecular , Ligação Proteica , Locos de Características Quantitativas , Receptores Imunológicos/química , Relação Estrutura-Atividade
9.
Cell Rep ; 22(3): 832-847, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29346778

RESUMO

Microglia, the CNS-resident immune cells, play important roles in disease, but the spectrum of their possible activation states is not well understood. We derived co-regulated gene modules from transcriptional profiles of CNS myeloid cells of diverse mouse models, including new tauopathy model datasets. Using these modules to interpret single-cell data from an Alzheimer's disease (AD) model, we identified microglial subsets-distinct from previously reported "disease-associated microglia"-expressing interferon-related or proliferation modules. We then analyzed whole-tissue RNA profiles from human neurodegenerative diseases, including a new AD dataset. Correcting for altered cellular composition of AD tissue, we observed elevated expression of the neurodegeneration-related modules, but also modules not implicated using expression profiles from mouse models alone. We provide a searchable, interactive database for exploring gene expression in all these datasets (http://research-pub.gene.com/BrainMyeloidLandscape). Understanding the dimensions of CNS myeloid cell activation in human disease may reveal opportunities for therapeutic intervention.


Assuntos
Doença de Alzheimer/genética , Encéfalo/metabolismo , Microglia/metabolismo , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos
10.
J Cell Biol ; 217(2): 459-472, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29196460

RESUMO

Proliferation and activation of microglia in the brain, concentrated around amyloid plaques, is a prominent feature of Alzheimer's disease (AD). Human genetics data point to a key role for microglia in the pathogenesis of AD. The majority of risk genes for AD are highly expressed (and many are selectively expressed) by microglia in the brain. There is mounting evidence that microglia protect against the incidence of AD, as impaired microglial activities and altered microglial responses to ß-amyloid are associated with increased AD risk. On the other hand, there is also abundant evidence that activated microglia can be harmful to neurons. Microglia can mediate synapse loss by engulfment of synapses, likely via a complement-dependent mechanism; they can also exacerbate tau pathology and secrete inflammatory factors that can injure neurons directly or via activation of neurotoxic astrocytes. Gene expression profiles indicate multiple states of microglial activation in neurodegenerative disease settings, which might explain the disparate roles of microglia in the development and progression of AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Microglia/metabolismo , Microglia/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos
11.
J Exp Med ; 214(9): 2611-2628, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28778989

RESUMO

Loss-of-function mutations in GRN cause frontotemporal dementia (FTD) with transactive response DNA-binding protein of 43 kD (TDP-43)-positive inclusions and neuronal ceroid lipofuscinosis (NCL). There are no disease-modifying therapies for either FTD or NCL, in part because of a poor understanding of how mutations in genes such as GRN contribute to disease pathogenesis and neurodegeneration. By studying mice lacking progranulin (PGRN), the protein encoded by GRN, we discovered multiple lines of evidence that PGRN deficiency results in impairment of autophagy, a key cellular degradation pathway. PGRN-deficient mice are sensitive to Listeria monocytogenes because of deficits in xenophagy, a specialized form of autophagy that mediates clearance of intracellular pathogens. Cells lacking PGRN display reduced autophagic flux, and pathological forms of TDP-43 typically cleared by autophagy accumulate more rapidly in PGRN-deficient neurons. Our findings implicate autophagy as a novel therapeutic target for GRN-associated NCL and FTD and highlight the emerging theme of defective autophagy in the broader FTD/amyotrophic lateral sclerosis spectrum of neurodegenerative disease.


Assuntos
Autofagia/fisiologia , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Animais , Granulinas , Listeria monocytogenes/imunologia , Listeriose/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Progranulinas , Transcriptoma
12.
Trends Mol Med ; 23(6): 512-533, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28442216

RESUMO

Alzheimer's disease (AD) is the most common form of dementia and the 6th leading cause of death in the US. The neuropathological hallmarks of the disease are extracellular amyloid-ß (Aß) plaques and intraneuronal hyperphosphorylated tau aggregates. Genetic variants of TREM2 (triggering receptor expressed on myeloid cells 2), a cell-surface receptor expressed selectively in myeloid cells, greatly increase the risk of AD, implicating microglia and the innate immune system as pivotal factors in AD pathogenesis. Recent studies have advanced our understanding of TREM2 biology and microglial activities in aging and neurodegenerative brains, providing new insights into TREM2 functions in amyloid plaque maintenance, microglial envelopment of plaque, microglia viability, and the identification of novel TREM2 ligands. Our increased understanding of TREM2 and microglia has opened new avenues for therapeutic intervention to delay or prevent the progression of AD.


Assuntos
Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Imunidade Inata , Glicoproteínas de Membrana/imunologia , Microglia/imunologia , Células Mieloides/imunologia , Receptores Imunológicos/imunologia , Envelhecimento/genética , Envelhecimento/patologia , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/imunologia , Animais , Humanos , Glicoproteínas de Membrana/genética , Microglia/patologia , Células Mieloides/patologia , Receptores Imunológicos/genética , Estados Unidos/epidemiologia
13.
J Alzheimers Dis ; 56(3): 1037-1054, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28106546

RESUMO

The common p.D358A variant (rs2228145) in IL-6R is associated with risk for multiple diseases and with increased levels of soluble IL-6R in the periphery and central nervous system (CNS). Here, we show that the p.D358A allele leads to increased proteolysis of membrane bound IL-6R and demonstrate that IL-6R peptides with A358 are more susceptible to cleavage by ADAM10 and ADAM17. IL-6 responsive genes were identified in primary astrocytes and microglia and an IL-6 gene signature was increased in the CNS of late onset Alzheimer's disease subjects in an IL6R allele dependent manner. We conducted a screen to identify variants associated with the age of onset of Alzheimer's disease in APOE ɛ4 carriers. Across five datasets, p.D358A had a meta P = 3 ×10-4 and an odds ratio = 1.3, 95% confidence interval 1.12 -1.48. Our study suggests that a common coding region variant of the IL-6 receptor results in neuroinflammatory changes that may influence the age of onset of Alzheimer's disease in APOE ɛ4 carriers.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Polimorfismo de Nucleotídeo Único , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Idoso , Idoso de 80 Anos ou mais , Alelos , Animais , Apolipoproteína E4/genética , Astrócitos/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Técnicas de Cocultura , Estudos de Coortes , Feminino , Células HEK293 , Humanos , Interleucina-6/metabolismo , Masculino , Camundongos , Microglia/metabolismo , Proteínas Recombinantes/metabolismo
14.
J Neurosci ; 36(38): 9962-75, 2016 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-27656033

RESUMO

UNLABELLED: After traumatic brain injury (TBI), neurons surviving the initial insult can undergo chronic (secondary) degeneration via poorly understood mechanisms, resulting in long-term cognitive impairment. Although a neuroinflammatory response is promptly activated after TBI, it is unknown whether it has a significant role in chronic phases of TBI (>1 year after injury). Using a closed-head injury model of TBI in mice, we showed by MRI scans that TBI caused substantial degeneration at the lesion site within a few weeks and these did not expand significantly thereafter. However, chronic alterations in neurons were observed, with reduced dendritic spine density lasting >1 year after injury. In parallel, we found a long-lasting inflammatory response throughout the entire brain. Deletion of one allele of CX3CR1, a chemokine receptor, limited infiltration of peripheral immune cells and largely prevented the chronic degeneration of the injured brain and provided a better functional recovery in female, but not male, mice. Therefore, targeting persistent neuroinflammation presents a new therapeutic option to reduce chronic neurodegeneration. SIGNIFICANCE STATEMENT: Traumatic brain injury (TBI) often causes chronic neurological problems including epilepsy, neuropsychiatric disorders, and dementia through unknown mechanisms. Our study demonstrates that inflammatory cells invading the brain lead to secondary brain damage. Sex-specific amelioration of chronic neuroinflammation rescues the brain degeneration and results in improved motor functions. Therefore, this study pinpoints an effective therapeutic approach to preventing secondary complications after TBI.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Inflamação/etiologia , Degeneração Neural , Recuperação de Função Fisiológica/fisiologia , Animais , Encéfalo/patologia , Receptor 1 de Quimiocina CX3C , Proteínas de Ligação ao Cálcio/metabolismo , Doença Crônica , Espinhas Dendríticas/imunologia , Espinhas Dendríticas/patologia , Espinhas Dendríticas/ultraestrutura , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Atividade Motora , Degeneração Neural/diagnóstico por imagem , Degeneração Neural/etiologia , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Desempenho Psicomotor/fisiologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Fatores de Tempo
15.
Cell Rep ; 16(6): 1690-1700, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27475227

RESUMO

The spread of tau pathology correlates with cognitive decline in Alzheimer's disease. In vitro, tau antibodies can block cell-to-cell tau spreading. Although mechanisms of anti-tau function in vivo are unknown, effector function might promote microglia-mediated clearance. In this study, we investigated whether antibody effector function is required for targeting tau. We compared efficacy in vivo and in vitro of two versions of the same tau antibody, with and without effector function, measuring tau pathology, neuron health, and microglial function. Both antibodies reduced accumulation of tau pathology in Tau-P301L transgenic mice and protected cultured neurons against extracellular tau-induced toxicity. Only the full-effector antibody enhanced tau uptake in cultured microglia, which promoted release of proinflammatory cytokines. In neuron-microglia co-cultures, only effectorless anti-tau protected neurons, suggesting full-effector tau antibodies can induce indirect toxicity via microglia. We conclude that effector function is not required for efficacy, and effectorless tau antibodies may represent a safer approach to targeting tau.


Assuntos
Doença de Alzheimer/metabolismo , Microglia/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Anticorpos/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Técnicas de Cocultura/métodos , Citocinas/metabolismo , Camundongos Transgênicos , Neurônios/metabolismo
16.
Nat Commun ; 7: 11295, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27097852

RESUMO

A common approach to understanding neurodegenerative disease is comparing gene expression in diseased versus healthy tissues. We illustrate that expression profiles derived from whole tissue RNA highly reflect the degenerating tissues' altered cellular composition, not necessarily transcriptional regulation. To accurately understand transcriptional changes that accompany neuropathology, we acutely purify neurons, astrocytes and microglia from single adult mouse brains and analyse their transcriptomes by RNA sequencing. Using peripheral endotoxemia to establish the method, we reveal highly specific transcriptional responses and altered RNA processing in each cell type, with Tnfr1 required for the astrocytic response. Extending the method to an Alzheimer's disease model, we confirm that transcriptomic changes observed in whole tissue are driven primarily by cell type composition, not transcriptional regulation, and identify hundreds of cell type-specific changes undetected in whole tissue RNA. Applying similar methods to additional models and patient tissues will transform our understanding of aberrant gene expression in neurological disease.


Assuntos
Doença de Alzheimer/genética , Astrócitos/metabolismo , Endotoxemia/genética , Microglia/metabolismo , Neurônios/metabolismo , Transcrição Gênica , Transcriptoma , Adulto , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Endotoxemia/metabolismo , Endotoxemia/patologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Especificidade de Órgãos , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Análise de Sequência de RNA
17.
Nat Med ; 20(12): 1452-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25419706

RESUMO

We have identified a rare coding mutation, T835M (rs137875858), in the UNC5C netrin receptor gene that segregated with disease in an autosomal dominant pattern in two families enriched for late-onset Alzheimer's disease and that was associated with disease across four large case-control cohorts (odds ratio = 2.15, Pmeta = 0.0095). T835M alters a conserved residue in the hinge region of UNC5C, and in vitro studies demonstrate that this mutation leads to increased cell death in human HEK293T cells and in rodent neurons. Furthermore, neurons expressing T835M UNC5C are more susceptible to cell death from multiple neurotoxic stimuli, including ß-amyloid (Aß), glutamate and staurosporine. On the basis of these data and the enriched hippocampal expression of UNC5C in the adult nervous system, we propose that one possible mechanism in which T835M UNC5C contributes to the risk of Alzheimer's disease is by increasing susceptibility to neuronal cell death, particularly in vulnerable regions of the Alzheimer's disease brain.


Assuntos
Doença de Alzheimer/genética , Neurônios/metabolismo , Receptores de Superfície Celular/genética , Receptores de Fator de Crescimento Neural/genética , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides , Animais , Região CA3 Hipocampal/citologia , Morte Celular/genética , Feminino , Predisposição Genética para Doença , Ácido Glutâmico , Células HEK293 , Humanos , Masculino , Camundongos , Receptores de Netrina , Ratos , Estaurosporina
18.
Nat Neurosci ; 16(11): 1576-87, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24097039

RESUMO

GABAergic cortical interneurons underlie the complexity of neural circuits and are particularly numerous and diverse in humans. In rodents, cortical interneurons originate in the subpallial ganglionic eminences, but their developmental origins in humans are controversial. We characterized the developing human ganglionic eminences and found that the subventricular zone (SVZ) expanded massively during the early second trimester, becoming densely populated with neural stem cells and intermediate progenitor cells. In contrast with the cortex, most stem cells in the ganglionic eminence SVZ did not maintain radial fibers or orientation. The medial ganglionic eminence exhibited unique patterns of progenitor cell organization and clustering, and markers revealed that the caudal ganglionic eminence generated a greater proportion of cortical interneurons in humans than in rodents. On the basis of labeling of newborn neurons in slice culture and mapping of proliferating interneuron progenitors, we conclude that the vast majority of human cortical interneurons are produced in the ganglionic eminences, including an enormous contribution from non-epithelial SVZ stem cells.


Assuntos
Ventrículos Cerebrais/citologia , Neurônios GABAérgicos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Interneurônios/fisiologia , Células-Tronco Multipotentes/fisiologia , Neocórtex/embriologia , Animais , Diferenciação Celular , Movimento Celular , Feto , Humanos , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Neocórtex/anatomia & histologia , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Cultura de Órgãos , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
19.
Nat Commun ; 4: 1665, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23575669

RESUMO

The human neocortex is increased in size and complexity as compared with most other species. Neocortical expansion has recently been attributed to protracted neurogenesis by outer radial glial cells in the outer subventricular zone, a region present in humans but not in rodents. The mechanisms of human outer radial glial cell generation are unknown, but are proposed to involve division of ventricular radial glial cells; neural stem cells present in all developing mammals. Here we show that human ventricular radial glial cells produce outer radial glial cells and seed formation of the outer subventricular zone via horizontal divisions, which occur more frequently in humans than in rodents. We further find that outer radial glial cell mitotic behaviour is cell intrinsic, and that the basal fibre, inherited by outer radial glial cells after ventricular radial glial division, determines cleavage angle. Our results suggest that altered regulation of mitotic spindle orientation increased outer radial glial cell number, and ultimately neuronal number, during human brain evolution.


Assuntos
Neocórtex/citologia , Neuroglia/citologia , Fuso Acromático , Humanos
20.
Cell ; 152(4): 895-908, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23375746

RESUMO

The mammalian telencephalon plays critical roles in cognition, motor function, and emotion. Though many of the genes required for its development have been identified, the distant-acting regulatory sequences orchestrating their in vivo expression are mostly unknown. Here, we describe a digital atlas of in vivo enhancers active in subregions of the developing telencephalon. We identified more than 4,600 candidate embryonic forebrain enhancers and studied the in vivo activity of 329 of these sequences in transgenic mouse embryos. We generated serial sets of histological brain sections for 145 reproducible forebrain enhancers, resulting in a publicly accessible web-based data collection comprising more than 32,000 sections. We also used epigenomic analysis of human and mouse cortex tissue to directly compare the genome-wide enhancer architecture in these species. These data provide a primary resource for investigating gene regulatory mechanisms of telencephalon development and enable studies of the role of distant-acting enhancers in neurodevelopmental disorders.


Assuntos
Elementos Facilitadores Genéticos , Telencéfalo/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Feto/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Telencéfalo/embriologia , Transcriptoma , Fatores de Transcrição de p300-CBP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA