Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 111: 32-45, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37004758

RESUMO

The molecular pathological mechanisms underlying schizophrenia remain unclear; however, genomic analysis has identified genes encoding important risk molecules. One such molecule is neurexin 1α (NRXN1α), a presynaptic cell adhesion molecule. In addition, novel autoantibodies that target the nervous system have been found in patients with encephalitis and neurological disorders. Some of these autoantibodies inhibit synaptic antigen molecules. Studies have examined the association between schizophrenia and autoimmunity; however, the pathological data remain unclear. Here, we identified a novel autoantibody against NRXN1α in patients with schizophrenia (n = 2.1%) in a Japanese cohort (n = 387). None of the healthy control participants (n = 362) were positive for anti-NRXN1α autoantibodies. Anti-NRXN1α autoantibodies isolated from patients with schizophrenia inhibited the molecular interaction between NRXN1α and Neuroligin 1 (NLGN1) and between NRXN1α and Neuroligin 2 (NLGN2). Additionally, these autoantibodies reduced the frequency of the miniature excitatory postsynaptic current in the frontal cortex of mice. Administration of anti-NRXN1α autoantibodies from patients with schizophrenia into the cerebrospinal fluid of mice reduced the number of spines/synapses in the frontal cortex and induced schizophrenia-related behaviors such as reduced cognition, impaired pre-pulse inhibition, and reduced social novelty preference. These changes were improved through the removal of anti-NRXN1α autoantibodies from the IgG fraction of patients with schizophrenia. These findings demonstrate that anti-NRXN1α autoantibodies transferred from patients with schizophrenia cause schizophrenia-related pathology in mice. Removal of anti-NRXN1α autoantibodies may be a therapeutic target for a subgroup of patients who are positive for these autoantibodies.


Assuntos
Esquizofrenia , Camundongos , Animais , Esquizofrenia/genética , Proteínas de Ligação ao Cálcio/metabolismo , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Autoanticorpos/metabolismo , Fenótipo
2.
Cell Rep Med ; 3(4): 100597, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35492247

RESUMO

From genetic and etiological studies, autoimmune mechanisms underlying schizophrenia are suspected; however, the details remain unclear. In this study, we describe autoantibodies against neural cell adhesion molecule (NCAM1) in patients with schizophrenia (5.4%, cell-based assay; 6.7%, ELISA) in a Japanese cohort (n = 223). Anti-NCAM1 autoantibody disrupts both NCAM1-NCAM1 and NCAM1-glial cell line-derived neurotrophic factor (GDNF) interactions. Furthermore, the anti-NCAM1 antibody purified from patients with schizophrenia interrupts NCAM1-Fyn interaction and inhibits phosphorylation of FAK, MEK1, and ERK1 when introduced into the cerebrospinal fluid of mice and also reduces the number of spines and synapses in frontal cortex. In addition, it induces schizophrenia-related behavior in mice, including deficient pre-pulse inhibition and cognitive impairment. In conclusion, anti-NCAM1 autoantibodies in patients with schizophrenia cause schizophrenia-related behavior and changes in synapses in mice. These antibodies may be a potential therapeutic target and serve as a biomarker to distinguish a small but treatable subgroup in heterogeneous patients with schizophrenia.


Assuntos
Moléculas de Adesão de Célula Nervosa , Esquizofrenia , Autoanticorpos , Antígeno CD56/genética , Humanos , Moléculas de Adesão de Célula Nervosa/genética , Esquizofrenia/genética , Sinapses/metabolismo
3.
Nat Commun ; 12(1): 6565, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34782623

RESUMO

Brain inflammation generally accompanies and accelerates neurodegeneration. Here we report a microglial mechanism in which polyglutamine binding protein 1 (PQBP1) senses extrinsic tau 3R/4R proteins by direct interaction and triggers an innate immune response by activating a cyclic GMP-AMP synthase (cGAS)-Stimulator of interferon genes (STING) pathway. Tamoxifen-inducible and microglia-specific depletion of PQBP1 in primary culture in vitro and mouse brain in vivo shows that PQBP1 is essential for sensing-tau to induce nuclear translocation of nuclear factor κB (NFκB), NFκB-dependent transcription of inflammation genes, brain inflammation in vivo, and eventually mouse cognitive impairment. Collectively, PQBP1 is an intracellular receptor in the cGAS-STING pathway not only for cDNA of human immunodeficiency virus (HIV) but also for the transmissible neurodegenerative disease protein tau. This study characterises a mechanism of brain inflammation that is common to virus infection and neurodegenerative disorders.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Encefalite/metabolismo , Proteínas de Membrana/metabolismo , Microglia/metabolismo , Nucleotidiltransferases/metabolismo , Animais , Encéfalo , Proteínas de Ligação a DNA/genética , Encefalite/imunologia , Feminino , HIV , Humanos , Imunidade Inata , Masculino , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , NF-kappa B/metabolismo , Doenças Neurodegenerativas , Nucleotidiltransferases/genética , Tamoxifeno/farmacologia
4.
Commun Biol ; 4(1): 1175, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34635772

RESUMO

DNA damage is increased in Alzheimer's disease (AD), while the underlying mechanisms are unknown. Here, we employ comprehensive phosphoproteome analysis, and identify abnormal phosphorylation of 70 kDa subunit of Ku antigen (Ku70) at Ser77/78, which prevents Ku70-DNA interaction, in human AD postmortem brains. The abnormal phosphorylation inhibits accumulation of Ku70 to the foci of DNA double strand break (DSB), impairs DNA damage repair and eventually causes transcriptional repression-induced atypical cell death (TRIAD). Cells under TRIAD necrosis reveal senescence phenotypes. Extracellular high mobility group box 1 (HMGB1) protein, which is released from necrotic or hyper-activated neurons in AD, binds to toll-like receptor 4 (TLR4) of neighboring neurons, and activates protein kinase C alpha (PKCα) that executes Ku70 phosphorylation at Ser77/78. Administration of human monoclonal anti-HMGB1 antibody to post-symptomatic AD model mice decreases neuronal DSBs, suppresses secondary TRIAD necrosis of neurons, prevents escalation of neurodegeneration, and ameliorates cognitive symptoms. TRIAD shares multiple features with senescence. These results discover the HMGB1-Ku70 axis that accounts for the increase of neuronal DNA damage and secondary enhancement of TRIAD, the cell death phenotype of senescence, in AD.


Assuntos
Doença de Alzheimer/patologia , Dano ao DNA , Reparo do DNA , Proteína HMGB1/fisiologia , Autoantígeno Ku/metabolismo , Transdução de Sinais/genética , Animais , Proteína HMGB1/genética , Camundongos , Camundongos Transgênicos , Fosforilação
5.
ACS Chem Neurosci ; 12(16): 3015-3027, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34319089

RESUMO

Tau aggregation is a central hallmark of tauopathies such as frontotemporal lobar degeneration and progressive supranuclear palsy as well as of Alzheimer's disease, and it has been a target for therapeutic development. Herein, we unexpectedly found that hepta-histidine (7H), an inhibitor of the interaction between Ku70 and Huntingtin proteins, suppresses aggregation of Tau-R3 peptides in vitro. Addition of the trans-activator of transcription (TAT) sequence (YGRKKRRQRRR) derived from the TAT protein to 7H increased its permeability into cells, and TAT-7H treatment of iPS cell-derived neurons carrying Tau or APP mutations suppressed Tau phosphorylation. These results indicate that 7H is a promising lead compound for developing anti-aggregation drugs against Tau-related neurodegenerative diseases including Alzheimer's disease (AD).


Assuntos
Doença de Alzheimer , Degeneração Lobar Frontotemporal , Tauopatias , Doença de Alzheimer/tratamento farmacológico , Histidina , Humanos , Proteínas tau
6.
Pharmaceuticals (Basel) ; 14(5)2021 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-34067842

RESUMO

CX3C Motif Chemokine Ligand 1 (CX3CL1; fractalkine) has been implicated in the pathogenesis of rheumatoid arthritis (RA) and its inhibition was found to attenuate arthritis in mice as well as in a clinical trial. Therefore, we investigated the effects of an anti-CX3CL1 monoclonal antibody (mAb) on immune-mediated interstitial lung disease (ILD) in SKG mice, which exhibit similar pathological and clinical features to human RA-ILD. CX3CL1 and CX3C chemokine receptor 1 (CX3CR1), the receptor for CX3CL1, were both expressed in the fibroblastic foci of lung tissue and the number of bronchoalveolar fluid (BALF) cells was elevated in ILD in SKG mice. No significant changes were observed in lung fibrosis or the number of BALF cells by the treatment with anti-CX3CL1 mAb. However, significantly greater reductions were observed in the number of M1 macrophages than in M2 macrophages in the BALF of treated mice. Furthermore, CX3CR1 expression levels were significantly higher in M1 macrophages than in M2 macrophages. These results suggest the stronger inhibitory effects of the anti-CX3CL1 mAb treatment against the alveolar infiltration of M1 macrophages than M2 macrophages in ILD in SKG mice. Thus, the CX3CL1-CX3CR1 axis may be involved in the infiltration of inflammatory M1 macrophages in RA-ILD.

7.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34130995

RESUMO

The early-stage pathologies of frontotemporal lobal degeneration (FTLD) remain largely unknown. In VCPT262A-KI mice carrying VCP gene mutation linked to FTLD, insufficient DNA damage repair in neural stem/progenitor cells (NSCs) activated DNA-PK and CDK1 that disabled MCM3 essential for the G1/S cell cycle transition. Abnormal neural exit produced neurons carrying over unrepaired DNA damage and induced early-stage transcriptional repression-induced atypical cell death (TRIAD) necrosis accompanied by the specific markers pSer46-MARCKS and YAP. In utero gene therapy expressing normal VCP or non-phosphorylated mutant MCM3 rescued DNA damage, neuronal necrosis, cognitive function, and TDP43 aggregation in adult neurons of VCPT262A-KI mice, whereas similar therapy in adulthood was less effective. The similar early-stage neuronal necrosis was detected in PGRNR504X-KI, CHMP2BQ165X-KI, and TDPN267S-KI mice, and blocked by embryonic treatment with AAV-non-phospho-MCM3. Moreover, YAP-dependent necrosis occurred in neurons of human FTLD patients, and consistently pSer46-MARCKS was increased in cerebrospinal fluid (CSF) and serum of these patients. Collectively, developmental stress followed by early-stage neuronal necrosis is a potential target for therapeutics and one of the earliest general biomarkers for FTLD.


Assuntos
Degeneração Lobar Frontotemporal/patologia , Células-Tronco Neurais/metabolismo , Proteína com Valosina/metabolismo , Animais , Ciclo Celular , Linhagem da Célula/genética , Células Cultivadas , Dano ao DNA/genética , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/líquido cefalorraquidiano , Degeneração Lobar Frontotemporal/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Necrose/metabolismo , Necrose/patologia , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Proteína com Valosina/genética
8.
Nat Commun ; 11(1): 507, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980612

RESUMO

The timing and characteristics of neuronal death in Alzheimer's disease (AD) remain largely unknown. Here we examine AD mouse models with an original marker, myristoylated alanine-rich C-kinase substrate phosphorylated at serine 46 (pSer46-MARCKS), and reveal an increase of neuronal necrosis during pre-symptomatic phase and a subsequent decrease during symptomatic phase. Postmortem brains of mild cognitive impairment (MCI) rather than symptomatic AD patients reveal a remarkable increase of necrosis. In vivo imaging reveals instability of endoplasmic reticulum (ER) in mouse AD models and genome-edited human AD iPS cell-derived neurons. The level of nuclear Yes-associated protein (YAP) is remarkably decreased in such neurons under AD pathology due to the sequestration into cytoplasmic amyloid beta (Aß) aggregates, supporting the feature of YAP-dependent necrosis. Suppression of early-stage neuronal death by AAV-YAPdeltaC reduces the later-stage extracellular Aß burden and cognitive impairment, suggesting that preclinical/prodromal YAP-dependent neuronal necrosis represents a target for AD therapeutics.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Proteínas de Ciclo Celular/metabolismo , Fatores de Transcrição/metabolismo , Doença de Alzheimer/líquido cefalorraquidiano , Peptídeos beta-Amiloides/metabolismo , Animais , Núcleo Celular/metabolismo , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/patologia , Simulação por Computador , Modelos Animais de Doenças , Retículo Endoplasmático/patologia , Retículo Endoplasmático/ultraestrutura , Feminino , Proteína HMGB1/líquido cefalorraquidiano , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lisofosfolipídeos/metabolismo , Masculino , Camundongos Transgênicos , Necrose , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Imagem com Lapso de Tempo , Proteínas de Sinalização YAP
9.
Methods Mol Biol ; 1880: 529-534, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30610719

RESUMO

Macroautophagy is the process to remove intracellular organelles or proteins by using autophagosome that is composed of autophagy proteins such as atg3, atg7, and atg8/LC3 (Mizushima, et al. Annu Rev Cell Dev Biol. 27:107-132, 2011). Here, we develop a useful method for in vivo imaging of autophagosome under the two-photon microscopy. Time-lapse imaging of LC3-ECFP enables us to quantify the dynamics of number, size, and signal intensity of autophagosomes in neurons or in other types of cells in the brain.


Assuntos
Autofagossomos/metabolismo , Autofagia/fisiologia , Encéfalo/diagnóstico por imagem , Microscopia Intravital/métodos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Animais , Proteínas Relacionadas à Autofagia/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Células HEK293 , Humanos , Processamento de Imagem Assistida por Computador/instrumentação , Processamento de Imagem Assistida por Computador/métodos , Microscopia Intravital/instrumentação , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Técnicas Estereotáxicas/instrumentação , Imagem com Lapso de Tempo/instrumentação , Imagem com Lapso de Tempo/métodos
10.
Mol Psychiatry ; 23(10): 2090-2110, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30283027

RESUMO

Early-phase pathologies of Alzheimer's disease (AD) are attracting much attention after clinical trials of drugs designed to remove beta-amyloid (Aß) aggregates failed to recover memory and cognitive function in symptomatic AD patients. Here, we show that phosphorylation of serine/arginine repetitive matrix 2 (SRRM2) at Ser1068, which is observed in the brains of early phase AD mouse models and postmortem end-stage AD patients, prevents its nuclear translocation by inhibiting interaction with T-complex protein subunit α. SRRM2 deficiency in neurons destabilized polyglutamine binding protein 1 (PQBP1), a causative gene for intellectual disability (ID), greatly affecting the splicing patterns of synapse-related genes, as demonstrated in a newly generated PQBP1-conditional knockout model. PQBP1 and SRRM2 were downregulated in cortical neurons of human AD patients and mouse AD models, and the AAV-PQBP1 vector recovered RNA splicing, the synapse phenotype, and the cognitive decline in the two mouse models. Finally, the kinases responsible for the phosphorylation of SRRM2 at Ser1068 were identified as ERK1/2 (MAPK3/1). These results collectively reveal a new aspect of AD pathology in which a phosphorylation signal affecting RNA splicing and synapse integrity precedes the formation of extracellular Aß aggregates and may progress in parallel with tau phosphorylation.


Assuntos
Doença de Alzheimer/genética , Proteínas de Transporte/genética , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Transporte Ativo do Núcleo Celular , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Cognição , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas , Deficiência Intelectual/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Cultura Primária de Células , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Proteínas tau/metabolismo
11.
eNeuro ; 5(4)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30225354

RESUMO

Phosphorylation of myristoylated alanine-rich C kinase substrate (MARCKS) reflects neurite degeneration at the early stage of Alzheimer's disease (AD), before extracellular Aß aggregates are histologically detectable. Here, we demonstrate that similar changes in MARCKS occur in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) pathologies in both mouse models and human patients. The increase in the level of pSer46-MARCKS began before α-synuclein aggregate formation, at a time when human α-Syn-BAC-Tg/GBA-hetero-KO mice exhibited no symptoms, and was sustained during aging, consistent with the pattern in human postmortem brains. The results strongly imply a common mechanism of pre-aggregation neurite degeneration in AD and PD/DLB pathologies.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer , Doença por Corpos de Lewy , Substrato Quinase C Rico em Alanina Miristoilada/metabolismo , Neuritos/patologia , Doença de Parkinson , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Masculino , Camundongos , Camundongos Transgênicos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fosforilação
12.
Hum Mol Genet ; 25(20): 4432-4447, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28173122

RESUMO

DNA damage and repair is a critical domain of many neurodegenerative diseases. In this study, we focused on RpA1, a candidate key molecule in polyQ disease pathologies, and tested the therapeutic effect of adeno-associated virus (AAV) vector expressing RpA1 on mutant Ataxin-1 knock-in (Atxn1-KI) mice. We found significant effects on motor functions, normalized DNA damage markers (γH2AX and 53BP1), and improved Purkinje cell morphology; effects that lasted for 50 weeks following AAV-RpA1 infection. In addition, we confirmed that AAV-RpA1 indirectly recovered multiple cellular functions such as RNA splicing, transcription and cell cycle as well as abnormal morphology of dendrite and dendritic spine of Purkinje cells in Atxn1-KI mice. All these results suggested a possibility of gene therapy with RpA1 for SCA1.


Assuntos
Ataxina-1/genética , Reparo do DNA , Mutação , Proteína de Replicação A/metabolismo , Ataxias Espinocerebelares/metabolismo , Animais , Ciclo Celular , DNA/metabolismo , Dano ao DNA , Dependovirus , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Terapia Genética , Camundongos , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Células de Purkinje/fisiologia , RNA/metabolismo , Splicing de RNA , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Ataxias Espinocerebelares/fisiopatologia , Transcrição Gênica
13.
Sci Rep ; 5: 12115, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26169250

RESUMO

We developed a new technique to observe macroautophagy in the brain in vivo, and examined whether fasting induced macroautophagy in neurons and how the induction was different between Alzheimer's disease (AD) model and control mice. Lentivirus for EGFP-LC3 injected into the brain successfully visualized autophagosome in living neurons by two-photon microscopy. The time-lapse imaging revealed that fasting increased the number, size and signal intensity of autophagosome in neurons. In AD model mice, these parameters of autophagosome were higher at the basal levels before starvation, and increased more rapidly by fasting than in control mice. However, metabolism of exogenous labeled Aß evaluated by the new technique suggested that the activated macroautophagy was insufficient to degrade the intracellular Aß increased by enhanced uptake from extracellular space after fasting. Ordinary immunohistochemistry also revealed that fasting increased intracellular accumulation of endogenous Aß, triggered cell dysfunction but did not mostly decrease extracellular Aß accumulation. Moreover, we unexpectedly discovered a circadian rhythm of basal level of macroautophagy. These results revealed new aspects of neuronal autophagy in normal/AD states and indicated usefulness of our method for evaluating autophagy functions in vivo.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Autofagia , Jejum/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Ritmo Circadiano , Modelos Animais de Doenças , Espaço Extracelular/metabolismo , Espaço Intracelular/metabolismo , Camundongos , Microscopia de Fluorescência , Neurônios/citologia , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA