Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Immunity ; 55(5): 879-894.e6, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35443157

RESUMO

The principal signals that drive memory and cognitive impairment in Alzheimer's disease (AD) remain elusive. Here, we revealed brain-wide cellular reactions to type I interferon (IFN-I), an innate immune cytokine aberrantly elicited by amyloid ß plaques, and examined their role in cognition and neuropathology relevant to AD in a murine amyloidosis model. Using a fate-mapping reporter system to track cellular responses to IFN-I, we detected robust, Aß-pathology-dependent IFN-I activation in microglia and other cell types. Long-term blockade of IFN-I receptor (IFNAR) rescued both memory and synaptic deficits and resulted in reduced microgliosis, inflammation, and neuritic pathology. Microglia-specific Ifnar1 deletion attenuated the loss of post-synaptic terminals by selective engulfment, whereas neural Ifnar1 deletion restored pre-synaptic terminals and decreased plaque accumulation. Overall, IFN-I signaling represents a critical module within the neuroinflammatory network of AD and prompts concerted cellular states that are detrimental to memory and cognition.


Assuntos
Doença de Alzheimer , Interferon Tipo I , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Imunidade Inata , Interferon Tipo I/metabolismo , Transtornos da Memória/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Placa Amiloide/metabolismo
2.
Cell ; 153(5): 1134-48, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23664764

RESUMO

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Epigenômica , Regulação da Expressão Gênica no Desenvolvimento , Animais , Diferenciação Celular , Cromatina/metabolismo , Ilhas de CpG , Células-Tronco Embrionárias/citologia , Histonas/metabolismo , Humanos , Metilação , Neoplasias/genética , Regiões Promotoras Genéticas , Peixe-Zebra/embriologia
3.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34187889

RESUMO

The autophagy-lysosomal pathway plays a critical role in intracellular clearance and metabolic homeostasis. While neuronal autophagy is known to participate in the degradation of neurofibrillary tangles composed of hyperphosphorylated and misfolded tau protein in Alzheimer's disease and other tauopathies, how microglial-specific autophagy regulates microglial intrinsic properties and neuronal tau pathology is not well understood. We report here that Atg7, a key mediator of autophagosome biogenesis, plays an essential role in the regulation of microglial lipid metabolism and neuroinflammation. Microglia-specific deletion of Atg7 leads to the transition of microglia to a proinflammatory status in vivo and to inflammasome activation in vitro. Activation of ApoE and lipid efflux attenuates the lipid droplets accumulation and inhibits cytokine production in microglial cells with Atg7 deficiency. Functionally, we show that the absence of microglial Atg7 enhances intraneuronal tau pathology and its spreading. Our results reveal an essential role for microglial autophagy in regulating lipid homeostasis, neuroinflammation, and tau pathology.


Assuntos
Autofagia , Homeostase , Metabolismo dos Lipídeos , Microglia/metabolismo , Tauopatias/patologia , Animais , Proteína 7 Relacionada à Autofagia/deficiência , Proteína 7 Relacionada à Autofagia/metabolismo , Linhagem Celular , Citocinas/metabolismo , Metabolismo Energético , Gliose/patologia , Inflamassomos/metabolismo , Inflamação/patologia , Camundongos Transgênicos
4.
Proc Natl Acad Sci U S A ; 112(40): 12516-21, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26392547

RESUMO

Human pluripotent stem cell-based in vitro models that reflect human physiology have the potential to reduce the number of drug failures in clinical trials and offer a cost-effective approach for assessing chemical safety. Here, human embryonic stem (ES) cell-derived neural progenitor cells, endothelial cells, mesenchymal stem cells, and microglia/macrophage precursors were combined on chemically defined polyethylene glycol hydrogels and cultured in serum-free medium to model cellular interactions within the developing brain. The precursors self-assembled into 3D neural constructs with diverse neuronal and glial populations, interconnected vascular networks, and ramified microglia. Replicate constructs were reproducible by RNA sequencing (RNA-Seq) and expressed neurogenesis, vasculature development, and microglia genes. Linear support vector machines were used to construct a predictive model from RNA-Seq data for 240 neural constructs treated with 34 toxic and 26 nontoxic chemicals. The predictive model was evaluated using two standard hold-out testing methods: a nearly unbiased leave-one-out cross-validation for the 60 training compounds and an unbiased blinded trial using a single hold-out set of 10 additional chemicals. The linear support vector produced an estimate for future data of 0.91 in the cross-validation experiment and correctly classified 9 of 10 chemicals in the blinded trial.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Neurais/citologia , Células-Tronco Pluripotentes/citologia , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Humanos , Hidrogéis/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Modelos Biológicos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Polietilenoglicóis/farmacologia , Máquina de Vetores de Suporte , Engenharia Tecidual/métodos , Xenobióticos/classificação , Xenobióticos/farmacologia
5.
Proc Natl Acad Sci U S A ; 110(39): 15644-9, 2013 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-23940360

RESUMO

Genome engineering in human pluripotent stem cells (hPSCs) holds great promise for biomedical research and regenerative medicine. Recently, an RNA-guided, DNA-cleaving interference pathway from bacteria [the type II clustered, regularly interspaced, short palindromic repeats (CRISPR)-CRISPR-associated (Cas) pathway] has been adapted for use in eukaryotic cells, greatly facilitating genome editing. Only two CRISPR-Cas systems (from Streptococcus pyogenes and Streptococcus thermophilus), each with their own distinct targeting requirements and limitations, have been developed for genome editing thus far. Furthermore, limited information exists about homology-directed repair (HDR)-mediated gene targeting using long donor DNA templates in hPSCs with these systems. Here, using a distinct CRISPR-Cas system from Neisseria meningitidis, we demonstrate efficient targeting of an endogenous gene in three hPSC lines using HDR. The Cas9 RNA-guided endonuclease from N. meningitidis (NmCas9) recognizes a 5'-NNNNGATT-3' protospacer adjacent motif (PAM) different from those recognized by Cas9 proteins from S. pyogenes and S. thermophilus (SpCas9 and StCas9, respectively). Similar to SpCas9, NmCas9 is able to use a single-guide RNA (sgRNA) to direct its activity. Because of its distinct protospacer adjacent motif, the N. meningitidis CRISPR-Cas machinery increases the sequence contexts amenable to RNA-directed genome editing.


Assuntos
Proteínas de Bactérias/metabolismo , Engenharia Genética/métodos , Genoma Humano/genética , Neisseria meningitidis/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Deleção de Genes , Marcação de Genes , Humanos , Mamíferos , Dados de Sequência Molecular , RNA/metabolismo , Edição de RNA/genética , Pequeno RNA não Traduzido
6.
Bioinformatics ; 30(18): 2665-7, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24872422

RESUMO

UNLABELLED: Aptamers are 'synthetic antibodies' that can bind to target molecules with high affinity and specificity. Aptamers are chemically synthesized and their discovery can be performed completely in vitro, rather than relying on in vivo biological processes, making them well-suited for high-throughput discovery. However, a large fraction of the most enriched aptamers in Systematic Evolution of Ligands by EXponential enrichment (SELEX) rounds display poor binding activity. Here, we present MPBind, a Meta-motif-based statistical framework and pipeline to Predict the BIND: ing potential of SELEX-derived aptamers. Using human embryonic stem cell SELEX-Seq data, MPBind achieved high prediction accuracy for binding potential. Further analysis showed that MPBind is robust to both polymerase chain reaction amplification bias and incomplete sequencing of aptamer pools. These two biases usually confound aptamer analysis. AVAILABILITY AND IMPLEMENTATION: MPBind software and documents are available at http://www.morgridge.net/MPBind.html. The human embryonic stem cells whole-cell SELEX-Seq data are available at http://www.morgridge.net/Aptamer/.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Biologia Computacional/métodos , Técnica de Seleção de Aptâmeros , Software , Células-Tronco Embrionárias/metabolismo , Humanos , Ligantes , Oligonucleotídeos/metabolismo , Análise de Sequência , Especificidade por Substrato
7.
Nat Methods ; 8(5): 424-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21478862

RESUMO

We re-examine the individual components for human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) culture and formulate a cell culture system in which all protein reagents for liquid media, attachment surfaces and splitting are chemically defined. A major improvement is the lack of a serum albumin component, as variations in either animal- or human-sourced albumin batches have previously plagued human ESC and iPSC culture with inconsistencies. Using this new medium (E8) and vitronectin-coated surfaces, we demonstrate improved derivation efficiencies of vector-free human iPSCs with an episomal approach. This simplified E8 medium should facilitate both the research use and clinical applications of human ESCs and iPSCs and their derivatives, and should be applicable to other reprogramming methods.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura/química , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Biópsia , Bovinos , Proliferação de Células , Sobrevivência Celular , Materiais Revestidos Biocompatíveis , Meios de Cultura Livres de Soro/química , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/citologia , Expressão Gênica , Substâncias de Crescimento , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariotipagem , Soroalbumina Bovina , Pele/citologia , Vitronectina
8.
J Clin Invest ; 133(12)2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37317973

RESUMO

Microglia are the major cell type expressing complement C3a receptor (C3aR) in the brain. Using a knockin mouse line in which a Td-tomato reporter is incorporated into the endogenous C3ar1 locus, we identified 2 major subpopulations of microglia with differential C3aR expression. Expressing the Td-tomato reporter on the APPNL-G-F-knockin (APP-KI) background revealed a significant shift of microglia to a high-C3aR-expressing subpopulation and they were enriched around amyloid ß (Aß) plaques. Transcriptomic analysis of C3aR-positive microglia documented dysfunctional metabolic signatures, including upregulation of hypoxia-inducible factor 1 (HIF-1) signaling and abnormal lipid metabolism in APP-KI mice compared with wild-type controls. Using primary microglial cultures, we found that C3ar1-null microglia had lower HIF-1α expression and were resistant to hypoxia mimetic-induced metabolic changes and lipid droplet accumulation. These were associated with improved receptor recycling and Aß phagocytosis. Crossing C3ar1-knockout mice with the APP-KI mice showed that C3aR ablation rescued the dysregulated lipid profiles and improved microglial phagocytic and clustering abilities. These were associated with ameliorated Aß pathology and restored synaptic and cognitive function. Our studies identify a heightened C3aR/HIF-1α signaling axis that influences microglial metabolic and lipid homeostasis in Alzheimer disease, suggesting that targeting this pathway may offer therapeutic benefit.


Assuntos
Peptídeos beta-Amiloides , Microglia , Animais , Camundongos , Peptídeos beta-Amiloides/genética , Receptores de Complemento , Camundongos Knockout , Lipídeos
9.
Sci Transl Med ; 14(648): eabj2658, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35675433

RESUMO

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic risk factors for Parkinson's disease (PD). Increased LRRK2 kinase activity is thought to impair lysosomal function and may contribute to the pathogenesis of PD. Thus, inhibition of LRRK2 is a potential disease-modifying therapeutic strategy for PD. DNL201 is an investigational, first-in-class, CNS-penetrant, selective, ATP-competitive, small-molecule LRRK2 kinase inhibitor. In preclinical models, DNL201 inhibited LRRK2 kinase activity as evidenced by reduced phosphorylation of both LRRK2 at serine-935 (pS935) and Rab10 at threonine-73 (pT73), a direct substrate of LRRK2. Inhibition of LRRK2 by DNL201 demonstrated improved lysosomal function in cellular models of disease, including primary mouse astrocytes and fibroblasts from patients with Gaucher disease. Chronic administration of DNL201 to cynomolgus macaques at pharmacologically relevant doses was not associated with adverse findings. In phase 1 and phase 1b clinical trials in 122 healthy volunteers and in 28 patients with PD, respectively, DNL201 at single and multiple doses inhibited LRRK2 and was well tolerated at doses demonstrating LRRK2 pathway engagement and alteration of downstream lysosomal biomarkers. Robust cerebrospinal fluid penetration of DNL201 was observed in both healthy volunteers and patients with PD. These data support the hypothesis that LRRK2 inhibition has the potential to correct lysosomal dysfunction in patients with PD at doses that are generally safe and well tolerated, warranting further clinical development of LRRK2 inhibitors as a therapeutic modality for PD.


Assuntos
Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Doença de Parkinson , Animais , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Lisossomos/metabolismo , Camundongos , Mutação , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Fosforilação
10.
Annu Rev Pathol ; 16: 277-298, 2021 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-33234021

RESUMO

Classic innate immune signaling pathways provide most of the immune response in the brain. This response activates many of the canonical signaling mechanisms identified in peripheral immune cells, despite their relative absence in this immune-privileged tissue. Studies over the past decade have strongly linked complement protein production and activation to age-related functional changes and neurodegeneration. The reactivation of the complement signaling pathway in aging and disease has opened new avenues for understanding brain aging and neurological disease pathogenesis and has implicated cell types such as astrocytes, microglia, endothelial cells, oligodendrocytes, neurons, and even peripheral immune cells in these processes. In this review, we aim to unravel the past decade of research related to complement activation and its numerous consequences in aging and neurological disease.


Assuntos
Envelhecimento/imunologia , Envelhecimento/patologia , Encéfalo/imunologia , Encéfalo/patologia , Proteínas do Sistema Complemento/fisiologia , Doenças Neurodegenerativas , Doença de Alzheimer/patologia , Animais , Humanos , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia
11.
J Clin Invest ; 131(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-32990682

RESUMO

Dysfunction of immune and vascular systems has been implicated in aging and Alzheimer disease; however, their interrelatedness remains poorly understood. The complement pathway is a well-established regulator of innate immunity in the brain. Here, we report robust age-dependent increases in vascular inflammation, peripheral lymphocyte infiltration, and blood-brain barrier (BBB) permeability. These phenotypes were subdued by global inactivation and by endothelial cell-specific ablation of C3ar1. Using an in vitro model of the BBB, we identified intracellular Ca2+ as a downstream effector of C3a/C3aR signaling and a functional mediator of vascular endothelial cadherin junction and barrier integrity. Endothelial C3ar1 inactivation also dampened microglia reactivity and improved hippocampal and cortical volumes in the aging brain, demonstrating a crosstalk between brain vasculature dysfunction and immune cell activation and neurodegeneration. Further, prominent C3aR-dependent vascular inflammation was also observed in a tau-transgenic mouse model. Our studies suggest that heightened C3a/C3aR signaling through endothelial cells promotes vascular inflammation and BBB dysfunction and contributes to overall neuroinflammation in aging and neurodegenerative disease.


Assuntos
Envelhecimento/metabolismo , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar , Endotélio Vascular/metabolismo , Receptores de Complemento/metabolismo , Vasculite/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Barreira Hematoencefálica/patologia , Complemento C3a/genética , Complemento C3a/metabolismo , Endotélio Vascular/patologia , Camundongos , Camundongos Knockout , Receptores de Complemento/genética , Vasculite/genética , Vasculite/patologia
12.
Sci Transl Med ; 12(573)2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298560

RESUMO

Neuroinflammation has been increasingly recognized to play a critical role in Alzheimer's disease (AD). The epoxy fatty acids (EpFAs) are derivatives of the arachidonic acid metabolism pathway and have anti-inflammatory activities. However, their efficacy is limited because of their rapid hydrolysis by the soluble epoxide hydrolase (sEH). We report that sEH is predominantly expressed in astrocytes and is elevated in postmortem brain tissue from patients with AD and in the 5xFAD ß amyloid mouse model of AD. The amount of sEH expressed in AD mouse brains correlated with a reduction in brain EpFA concentrations. Using a specific small-molecule sEH inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), we report that TPPU treatment protected wild-type mice against LPS-induced inflammation in vivo. Long-term administration of TPPU to the 5xFAD mouse model via drinking water reversed microglia and astrocyte reactivity and immune pathway dysregulation. This was associated with reduced ß amyloid pathology and improved synaptic integrity and cognitive function on two behavioral tests. TPPU treatment correlated with an increase in EpFA concentrations in the brains of 5xFAD mice, demonstrating brain penetration and target engagement of this small molecule. These findings support further investigation of TPPU as a potential therapeutic agent for the treatment of AD.


Assuntos
Doença de Alzheimer , Epóxido Hidrolases , Doença de Alzheimer/tratamento farmacológico , Animais , Compostos de Epóxi , Humanos , Camundongos , Compostos de Fenilureia , Piperidinas
13.
J Clin Invest ; 130(4): 1912-1930, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31917687

RESUMO

Type I interferon (IFN) is a key cytokine that curbs viral infection and cell malignancy. Previously, we demonstrated a potent IFN immunogenicity of nucleic acid-containing (NA-containing) amyloid fibrils in the periphery. Here, we investigated whether IFN is associated with ß-amyloidosis inside the brain and contributes to neuropathology. An IFN-stimulated gene (ISG) signature was detected in the brains of multiple murine Alzheimer disease (AD) models, a phenomenon also observed in WT mouse brain challenged with generic NA-containing amyloid fibrils. In vitro, microglia innately responded to NA-containing amyloid fibrils. In AD models, activated ISG-expressing microglia exclusively surrounded NA+ amyloid ß plaques, which accumulated in an age-dependent manner. Brain administration of rIFN-ß resulted in microglial activation and complement C3-dependent synapse elimination in vivo. Conversely, selective IFN receptor blockade effectively diminished the ongoing microgliosis and synapse loss in AD models. Moreover, we detected activated ISG-expressing microglia enveloping NA-containing neuritic plaques in postmortem brains of patients with AD. Gene expression interrogation revealed that IFN pathway was grossly upregulated in clinical AD and significantly correlated with disease severity and complement activation. Therefore, IFN constitutes a pivotal element within the neuroinflammatory network of AD and critically contributes to neuropathogenic processes.


Assuntos
Doença de Alzheimer/imunologia , Amiloide/imunologia , Interferon beta/imunologia , Sinapses/imunologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Animais , Complemento C3/imunologia , Modelos Animais de Doenças , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Interferon beta/efeitos adversos , Interferon beta/farmacologia , Camundongos , Microglia/imunologia , Microglia/patologia , Sinapses/patologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
14.
JCI Insight ; 3(13)2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-29997299

RESUMO

Nonneuronal cell types in the CNS are increasingly implicated as critical players in brain health and disease. While gene expression profiling of bulk brain tissue is routinely used to examine alterations in the brain under various conditions, it does not capture changes that occur within single cell types or allow interrogation of crosstalk among cell types. To this end, we have developed a concurrent brain cell type acquisition (CoBrA) methodology, enabling the isolation and profiling of microglia, astrocytes, endothelia, and oligodendrocytes from a single adult mouse forebrain. By identifying and validating anti-ACSA-2 and anti-CD49a antibodies as cell surface markers for astrocytes and vascular endothelial cells, respectively, and using established antibodies to isolate microglia and oligodendrocytes, we document that these 4 major cell types are isolated with high purity and RNA quality. We validated our procedure by performing acute peripheral LPS challenge, while highlighting the underappreciated changes occurring in astrocytes and vascular endothelia in addition to microglia. Furthermore, we assessed cell type-specific gene expression changes in response to amyloid pathology in a mouse model of Alzheimer's disease. Our CoBrA methodology can be readily implemented to interrogate multiple CNS cell types in any mouse model at any age.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Inflamação/patologia , Doença de Alzheimer/genética , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Inflamação/genética , Integrina alfa1 , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia
15.
Neuron ; 100(6): 1337-1353.e5, 2018 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-30415998

RESUMO

Strong evidence implicates the complement pathway as an important contributor to amyloid pathology in Alzheimer's disease (AD); however, the role of complement in tau modulation remains unclear. Here we show that the expression of C3 and C3a receptor (C3aR1) are positively correlated with cognitive decline and Braak staging in human AD brains. Deletion of C3ar1 in PS19 mice results in the rescue of tau pathology and attenuation of neuroinflammation, synaptic deficits, and neurodegeneration. Through RNA sequencing and cell-type-specific transcriptomic analysis, we identify a C3aR-dependent transcription factor network that regulates a reactive glial switch whose inactivation ameliorates disease-associated microglia and neurotoxic astrocyte signatures. Strikingly, this C3aR network includes multiple genes linked to late-onset AD. Mechanistically, we identify STAT3 as a direct target of C3-C3aR signaling that functionally mediates tau pathogenesis. All together our findings demonstrate a crucial role for activation of the C3-C3aR network in mediating neuroinflammation and tau pathology.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/patologia , Complemento C3a/metabolismo , Citocinas/metabolismo , Receptores de Complemento/metabolismo , Transdução de Sinais/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Animais , Encéfalo/fisiopatologia , Proteínas de Ligação ao Cálcio , Transtornos Cognitivos/etiologia , Complemento C3a/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Potenciais Evocados/fisiologia , Feminino , Redes Reguladoras de Genes/fisiologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Receptores de Complemento/genética , Transdução de Sinais/efeitos dos fármacos , Tauopatias/complicações , Regulação para Cima/fisiologia , Proteínas tau/genética , Proteínas tau/metabolismo
16.
Exp Biol Med (Maywood) ; 242(17): 1679-1689, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28599598

RESUMO

The aim of the present study was to test sample reproducibility for model neural tissues formed on synthetic hydrogels. Human embryonic stem (ES) cell-derived precursor cells were cultured on synthetic poly(ethylene glycol) (PEG) hydrogels to promote differentiation and self-organization into model neural tissue constructs. Neural progenitor, vascular, and microglial precursor cells were combined on PEG hydrogels to mimic developmental timing, which produced multicomponent neural constructs with 3D neuronal and glial organization, organized vascular networks, and microglia with ramified morphologies. Spearman's rank correlation analysis of global gene expression profiles and a comparison of coefficient of variation for expressed genes demonstrated that replicate neural constructs were highly uniform to at least day 21 for samples from independent experiments. We also demonstrate that model neural tissues formed on PEG hydrogels using a simplified neural differentiation protocol correlated more strongly to in vivo brain development than samples cultured on tissue culture polystyrene surfaces alone. These results provide a proof-of-concept demonstration that 3D cellular models that mimic aspects of human brain development can be produced from human pluripotent stem cells with high sample uniformity between experiments by using standard culture techniques, cryopreserved cell stocks, and a synthetic extracellular matrix. Impact statement Pluripotent stem (PS) cells have been characterized by an inherent ability to self-organize into 3D "organoids" resembling stomach, intestine, liver, kidney, and brain tissues, offering a potentially powerful tool for modeling human development and disease. However, organoid formation must be quantitatively reproducible for applications such as drug and toxicity screening. Here, we report a strategy to produce uniform neural tissue constructs with reproducible global gene expression profiles for replicate samples from multiple experiments.


Assuntos
Encéfalo/citologia , Modelos Biológicos , Células-Tronco Neurais/citologia , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Hidrogéis , Polietilenoglicóis
17.
PLoS One ; 10(9): e0139076, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26406898

RESUMO

Growth factors and transcription factors are well known to regulate pluripotent stem cells, but less is known about translational control in stem cells. Here, we use embryonic stem cells (ESCs) to investigate a connection between ESC growth factors and eIF2α-mediated translational control (eIF2α phosphorylation promotes protein expression from mRNAs with upstream open-reading frames, or uORFs). We find abundant phosphorylated P-eIF2α (P-eIF2α) in both pluripotent mouse and human ESCs, but little P-eIF2α in ESCs triggered to differentiate. We show that the growth factors LIF (leukemia inhibitory factor) and BMP4 (bone morphogenic protein 4) both maintain P-eIF2α in mESCs, but use distinct mechanisms: LIF inhibits an eIF2α phosphatase whereas BMP4 activates an eIF2α kinase. The mRNAs encoding the pluripotency factors Nanog and c-Myc possess uORFs while Oct4 mRNA does not. We find that salubrinal, a chemical that increases eIF2α phosphorylation, promotes Nanog and c-Myc expression, but not Oct4 expression. These experiments connect ESC growth factors to eIF2α phosphorylation and suggest a chemical substitute for LIF to enhance Nanog and c-Myc expression.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Fator Inibidor de Leucemia/farmacologia , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Linhagem Celular , Cinamatos/farmacologia , Fator de Iniciação Eucariótico 4G/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Camundongos , Modelos Biológicos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Proteína Homeobox Nanog , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Tioureia/análogos & derivados , Tioureia/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA