Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Brain ; 144(5): 1526-1541, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34148071

RESUMO

Alzheimer's disease, characterized by brain deposits of amyloid-ß plaques and neurofibrillary tangles, is also linked to neurovascular dysfunction and blood-brain barrier breakdown, affecting the passage of substances into and out of the brain. We hypothesized that treatment of neurovascular alterations could be beneficial in Alzheimer's disease. Annexin A1 (ANXA1) is a mediator of glucocorticoid anti-inflammatory action that can suppress microglial activation and reduce blood-brain barrier leakage. We have reported recently that treatment with recombinant human ANXA1 (hrANXA1) reduced amyloid-ß levels by increased degradation in neuroblastoma cells and phagocytosis by microglia. Here, we show the beneficial effects of hrANXA1 in vivo by restoring efficient blood-brain barrier function and decreasing amyloid-ß and tau pathology in 5xFAD mice and Tau-P301L mice. We demonstrate that young 5xFAD mice already suffer cerebrovascular damage, while acute pre-administration of hrANXA1 rescued the vascular defects. Interestingly, the ameliorated blood-brain barrier permeability in young 5xFAD mice by hrANXA1 correlated with reduced brain amyloid-ß load, due to increased clearance and degradation of amyloid-ß by insulin degrading enzyme (IDE). The systemic anti-inflammatory properties of hrANXA1 were also observed in 5xFAD mice, increasing IL-10 and reducing TNF-α expression. Additionally, the prolonged treatment with hrANXA1 reduced the memory deficits and increased synaptic density in young 5xFAD mice. Similarly, in Tau-P301L mice, acute hrANXA1 administration restored vascular architecture integrity, affecting the distribution of tight junctions, and reduced tau phosphorylation. The combined data support the hypothesis that blood-brain barrier breakdown early in Alzheimer's disease can be restored by hrANXA1 as a potential therapeutic approach.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Anexina A1/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Animais , Barreira Hematoencefálica/patologia , Encéfalo/patologia , Permeabilidade Capilar , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos
2.
EBioMedicine ; 39: 377-387, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30502054

RESUMO

BACKGROUND: Glycogen synthase kinase-3ß (GSK3ß) is a key regulator of cellular homeostasis. In neurons, GSK3ß contributes to the control of neuronal transmission and plasticity, but its role in epilepsy remains to be defined. METHODS: Biochemical and electrophysiological methods were used to assess the role of GSK3ß in regulating neuronal transmission and epileptogenesis. GSK3ß activity was increased genetically in GSK3ß[S9A] mice. Its effects on neuronal transmission and epileptogenesis induced by kainic acid were assessed by field potential recordings in mice brain slices and video electroencephalography in vivo. The ion channel expression was measured in brain samples from mice and followed by analysis in samples from patients with temporal lobe epilepsy or focal cortical dysplasia in correlation to GSK3ß phosphorylation. FINDINGS: Higher GSK3ß activity decreased the progression of kainic acid induced epileptogenesis. At the biochemical level, higher GSK3ß activity increased the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel 4 under basal conditions and in the epileptic mouse brain and decreased phosphorylation of the glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 at Serine 831 under basal conditions. Moreover, we found a significant correlation between higher inhibitory GSK3ß phosphorylation at Serine 9 and higher activating GluA1 phosphorylation at Serine 845 in brain samples from epileptic patients. INTERPRETATION: Our data imply GSK3ß activity in the protection of neuronal networks from hyper-activation in response to epileptogenic stimuli and indicate that the anti-epileptogenic function of GSK3ß involves modulation of HCN4 level and the synaptic AMPA receptors pool.


Assuntos
Epilepsia/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Ácido Caínico/efeitos adversos , Proteínas Musculares/metabolismo , Canais de Potássio/metabolismo , Receptores de AMPA/metabolismo , Adolescente , Adulto , Animais , Células Cultivadas , Criança , Pré-Escolar , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia/induzido quimicamente , Epilepsia/genética , Feminino , Glicogênio Sintase Quinase 3 beta/química , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fosforilação , Receptores de AMPA/química , Transdução de Sinais , Transmissão Sináptica , Gravação em Vídeo
3.
Mol Neurobiol ; 55(7): 6050-6062, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29143288

RESUMO

Glycogen synthase kinases-3ß (GSK3ß) is a key regulator of cell homeostasis. In neurons, GSK3ß contributes to control of neuronal transmission and plasticity. Despite extensive studies in non-neuronal cells, crosstalk between GSK3ß and other signaling pathways remains not well defined in neurons. In the present study, we report that GSK3ß positively affected the activity of effectors of mammalian target of rapamycin complex 1 (mTORC1) and complex 2 (mTORC2), in mature neurons in vitro and in vivo. GSK3ß also promoted prosurvival signaling and attenuated kainic acid-induced apoptosis. Our study identified GSK3ß as a positive regulator of prosurvival signaling, including the mTOR pathway, and indicates the possible neuroprotective role of GSK3ß in models of pharmacologically induced excitotoxicity.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Neurônios/citologia , Neurônios/enzimologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Apoptose , Encéfalo/enzimologia , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Isoenzimas/metabolismo , Ácido Caínico , Camundongos Transgênicos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína S6 Ribossômica/metabolismo
4.
Brain ; 137(Pt 1): 78-91, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24277722

RESUMO

The chemokine fractalkine modulates microglial responses in neurodegenerative diseases, including tauopathies, but the mechanistic processes and their relevance in human brain pathologies is not yet known. Here, we show that hippocampal HT22 cells expressing human TAU(P301L) mutant protein produce fractalkine, which in microglia activates AKT, inhibits glycogen synthase kinase-3ß and upregulates the transcription factor NRF2/NFE2L2 and its target genes including heme oxygenase 1. In a mouse model of tauopathy based on stereotaxic delivery in hippocampus of an adeno-associated viral vector for expression of TAU(P301L), we confirmed that tau-injured neurons express fractalkine. NRF2- and fractalkine receptor-knockout mice did not express heme oxygenase 1 in microglia and exhibited increased microgliosis and astrogliosis in response to neuronal TAU(P301L) expression, demonstrating a crucial role of the fractalkine/NRF2/heme oxygenase 1 pathway in attenuation of the pro-inflammatory phenotype. The hippocampus of patients with Alzheimer's disease also exhibits increased expression of fractalkine in TAU-injured neurons that recruit microglia. These events correlated with increased levels of NRF2 and heme oxygenase 1 proteins, suggesting an attempt of the diseased brain to limit microgliosis. Our combined results indicate that fractalkine mobilizes NRF2 to limit over-activation of microglia and identify this new target to control unremitting neuroinflammation in tauopathies.


Assuntos
Quimiocina CX3CL1/farmacologia , Gliose/tratamento farmacológico , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Tauopatias/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Western Blotting , Encéfalo/patologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Feminino , Imunofluorescência , Gliose/etiologia , Gliose/patologia , Humanos , Imuno-Histoquímica , Inflamação/patologia , Lentivirus/genética , Masculino , Camundongos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Tauopatias/complicações , Tauopatias/patologia
5.
PLoS One ; 8(8): e72301, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977276

RESUMO

Progressive aggregation of protein Tau into oligomers and fibrils correlates with cognitive decline and synaptic dysfunction, leading to neurodegeneration in vulnerable brain regions in Alzheimer's disease. The unmet need of effective therapy for Alzheimer's disease, combined with problematic pharmacological approaches, led the field to explore immunotherapy, first against amyloid peptides and recently against protein Tau. Here we adapted the liposome-based amyloid vaccine that proved safe and efficacious, and incorporated a synthetic phosphorylated peptide to mimic the important phospho-epitope of protein Tau at residues pS396/pS404. We demonstrate that the liposome-based vaccine elicited, rapidly and robustly, specific antisera in wild-type mice and in Tau.P301L mice. Long-term vaccination proved to be safe, because it improved the clinical condition and reduced indices of tauopathy in the brain of the Tau.P301L mice, while no signs of neuro-inflammation or other adverse neurological effects were observed. The data corroborate the hypothesis that liposomes carrying phosphorylated peptides of protein Tau have considerable potential as safe and effective treatment against tauopathies, including Alzheimer's disease.


Assuntos
Vacinas contra Alzheimer/imunologia , Anticorpos Neutralizantes/sangue , Peptídeos/imunologia , Fosfoproteínas/imunologia , Tauopatias/tratamento farmacológico , Proteínas tau/imunologia , Vacinas contra Alzheimer/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Humanos , Lipossomos/química , Camundongos , Camundongos Transgênicos , Peptídeos/administração & dosagem , Peptídeos/síntese química , Fosfoproteínas/administração & dosagem , Fosfoproteínas/síntese química , Fosforilação , Desempenho Psicomotor/efeitos dos fármacos , Tauopatias/imunologia , Tauopatias/fisiopatologia , Resultado do Tratamento , Vacinação , Proteínas tau/antagonistas & inibidores , Proteínas tau/genética
6.
PLoS One ; 8(5): e63589, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23704923

RESUMO

Cell adhesion molecules are important structural substrates, required for synaptic plasticity and synaptogenesis. CAMs differ widely in their expression throughout different brain regions and their specific structural and functional roles in the brain remain to be elucidated. Here, we investigated selected cell adhesion molecules for alterations in expression levels and neuronal localization in validated mouse models for Alzheimer's disease that mimic the age-related progression of amyloid accumulation and tauopathy. Among the cell adhesion molecules analyzed, Nectin-3 expression was affected most and specifically in all mouse models with tauopathy. In particular was Nectin-3 depleted from the specific region of the hippocampus, known as the stratum lacunosum and moleculare, in mice that express wild-type or mutant human protein Tau, either chronically or sub-acutely. Tauopathy progresses from the entorhinal cortex to the hippocampus by unknown mechanisms that could involve transport by the myelinated axons of the temporoammonic and perforant pathways. The decreased expression of Nectin-3 in the stratum lacunosum moleculare is an early marker of impaired transport, and eventual synaptic problems, caused by beginning tauopathy.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Moléculas de Adesão Celular/metabolismo , Regulação para Baixo/genética , Tauopatias/metabolismo , Animais , Moléculas de Adesão Celular/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Nectinas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinaptossomos/metabolismo , Tauopatias/patologia
7.
Mol Cell Biol ; 32(21): 4410-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22927642

RESUMO

Alzheimer's disease (AD) has been associated with altered activity of glycogen synthase kinase 3 (GSK3) isozymes, which are proposed to contribute to both neurofibrillary tangles and amyloid plaque formation. However, the molecular basis by which GSK3 affects the formation of Aß remains unknown. Our aim was to identify the underlying mechanisms of GSK3-dependent effects on the processing of amyloid precursor protein (APP). For this purpose, N2a cells stably expressing APP carrying the Swedish mutation were treated with specific GSK3 inhibitors or transfected with GSK3α/ß short interfering RNA. We show that inhibition of GSK3 leads to decreased expression of APP by enhancing its degradation via an increase in the number of lysosomes. This induction of the lysosomal/autophagy pathway was associated with nuclear translocation of transcription factor EB (TFEB), a master regulator of lysosomal biogenesis. Our data indicate that GSK3 inhibition reduces Aß through an increase of the degradation of APP and its carboxy-terminal fragment (CTF) by activation of the lysosomal/autophagy pathway. These results suggest that an increased propensity toward autophagic/lysosomal alterations in AD patients could have consequences for neuronal function.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Autofagia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Lisossomos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/imunologia , Animais , Anticorpos Monoclonais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/imunologia , Células HEK293 , Humanos , Camundongos , Placa Amiloide , Interferência de RNA , RNA Interferente Pequeno
8.
J Med Chem ; 55(9): 4407-24, 2012 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-22533818

RESUMO

The glycogen synthase kinase-3 (GSK-3) has been linked to the pathogenesis of colorectal cancer, diabetes, cardiovascular disease, acute myeloid leukemia (AML), and Alzheimer's disease (AD). The debate on the respective contributions of GSK-3α and GSK-3ß to AD pathology and AML is ongoing. Thus, the identification of potent GSK-3α-selective inhibitors, endowed with favorable pharmacokinetic properties, may elucidate the effect of GSK-3α inhibition in AD and AML models. The analysis of all available crystallized GSK-3 structures provided a simplified scheme of the relevant hot spots responsible for ligand binding and potency. This resulted in the identification of novel scorpion shaped GSK-3 inhibitors. It is noteworthy, compounds 14d and 15b showed the highest GSK-3α selectivity reported so far. In addition, compound 14d did not display significant inhibition of 48 out of 50 kinases in the test panel. The GSK-3 inhibitors were further profiled for efficacy and toxicity in the wild-type (wt) zebrafish embryo assay.


Assuntos
Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Oxidiazóis/química , Oxidiazóis/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Modelos Moleculares , Simulação de Dinâmica Molecular , Oxidiazóis/síntese química , Inibidores de Proteínas Quinases/síntese química , Relação Estrutura-Atividade , Peixe-Zebra
9.
Am J Pathol ; 179(4): 2001-15, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21839061

RESUMO

Adeno-associated virus (AAV)-mediated expression of wild-type or mutant P301L protein tau produces massive degeneration of pyramidal neurons without protein tau aggregation. We probed this novel model for genetic and structural factors and early parameters of pyramidal neurodegeneration. In yellow fluorescent protein-expressing transgenic mice, intracerebral injection of AAV-tauP301L revealed early damage to apical dendrites of CA1 pyramidal neurons, whereas their somata remained normal. Ultrastructurally, more and enlarged autophagic vacuoles were contained in degenerating dendrites and manifested as dark, discontinuous, vacuolated processes surrounded by activated astrocytes. Dendritic spines were lost in AAV-tauP301L-injected yellow fluorescent protein-expressing transgenic mice, and ultrastructurally, spines appeared dark and degenerating. In CX3CR1(EGFP/EGFP)-deficient mice, microglia were recruited early to neurons expressing human tau. The inflammatory response was accompanied by extravasation of plasma immunoglobulins. α2-Macroglobulin, but neither albumin nor transferrin, became lodged in the brain parenchyma. Large proteins, but not Evans blue, entered the brain of mice injected with AAV-tauP301L. Ultrastructurally, brain capillaries were constricted and surrounded by swollen astrocytes with extensions that contacted degenerating dendrites and axons. Together, these data corroborate the hypothesis that neuroinflammation participates essentially in tau-mediated neurodegeneration, and the model recapitulates early dendritic defects reminiscent of "dendritic amputation" in Alzheimer's disease.


Assuntos
Dendritos/patologia , Inflamação/patologia , Degeneração Neural/patologia , Sistema Nervoso/irrigação sanguínea , Sistema Nervoso/patologia , Proteínas tau/metabolismo , Animais , Axônios/patologia , Axônios/ultraestrutura , Biomarcadores/metabolismo , Vasos Sanguíneos/patologia , Vasos Sanguíneos/ultraestrutura , Barreira Hematoencefálica/patologia , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/ultraestrutura , Dendritos/ultraestrutura , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/complicações , Masculino , Camundongos , Degeneração Neural/complicações , Estresse Oxidativo , Permeabilidade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Sinapses/patologia , Sinapses/ultraestrutura
10.
FASEB J ; 25(9): 3208-18, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21593432

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) has neuroprotective and neurotrophic properties and is a potent α-secretase activator. As PACAP peptides and their specific receptor PAC1 are localized in central nervous system areas affected by Alzheimer's disease (AD), this study aims to examine the role of the natural peptide PACAP as a valuable approach in AD therapy. We investigated the effect of PACAP in the brain of an AD transgenic mouse model. The long-term intranasal daily PACAP application stimulated the nonamyloidogenic processing of amyloid precursor protein (APP) and increased expression of the brain-derived neurotrophic factor and of the antiapoptotic Bcl-2 protein. In addition, it caused a strong reduction of the amyloid ß-peptide (Aß) transporter receptor for advanced glycation end products (RAGE) mRNA level. PACAP, by activation of the somatostatin-neprilysin cascade, also enhanced expression of the Aß-degrading enzyme neprilysin in the mouse brain. Furthermore, daily PAC1-receptor activation via PACAP resulted in an increased mRNA level of both the PAC1 receptor and its ligand PACAP. Our behavioral studies showed that long-term PACAP treatment of APP[V717I]-transgenic mice improved cognitive function in animals. Thus, nasal application of PACAP was effective, and our results indicate that PACAP could be of therapeutic value in treating AD.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Administração Intranasal , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neprilisina/genética , Neprilisina/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(17): 6957-62, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21482799

RESUMO

Functional diversity of protein phosphatase 2A (PP2A) enzymes mainly results from their association with distinct regulatory subunits. To analyze the functions of one such holoenzyme in vivo, we generated mice lacking PR61/B'δ (B56δ), a subunit highly expressed in neural tissues. In PR61/B'δ-null mice the microtubule-associated protein tau becomes progressively phosphorylated at pathological epitopes in restricted brain areas, with marked immunoreactivity for the misfolded MC1-conformation but without neurofibrillary tangle formation. Behavioral tests indicated impaired sensorimotor but normal cognitive functions. These phenotypical characteristics were further underscored in PR61/B'δ-null mice mildly overexpressing human tau. PR61/B'δ-containing PP2A (PP2A(T61δ)) poorly dephosphorylates tau in vitro, arguing against a direct dephosphorylation defect. Rather, the activity of glycogen synthase kinase-3ß, a major tau kinase, was found increased, with decreased phosphorylation of Ser-9, a putative cyclin-dependent kinase 5 (CDK5) target. Accordingly, CDK5 activity is decreased, and its cellular activator p35, strikingly absent in the affected brain areas. As opposed to tau, p35 is an excellent PP2A(T61δ) substrate. Our data imply a nonredundant function for PR61/B'δ in phospho-tau homeostasis via an unexpected spatially restricted mechanism preventing p35 hyperphosphorylation and its subsequent degradation.


Assuntos
Encéfalo/enzimologia , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Dobramento de Proteína , Proteína Fosfatase 2/metabolismo , Tauopatias/enzimologia , Animais , Quinase 5 Dependente de Ciclina/genética , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células PC12 , Fosforilação/genética , Proteína Fosfatase 2/genética , Ratos , Tauopatias/genética , Proteínas tau/genética , Proteínas tau/metabolismo
12.
Curr Biol ; 21(4): 316-21, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21315593

RESUMO

Mustard oil (MO) is a plant-derived irritant that has been extensively used in experimental models to induce pain and inflammation. The noxious effects of MO are currently ascribed to specific activation of the cation channel TRPA1 in nociceptive neurons. In contrast to this view, we show here that the capsaicin receptor TRPV1 has a surprisingly large contribution to aversive and pain responses and visceral irritation induced by MO. Furthermore, we found that this can be explained by previously unknown properties of this compound. First, MO has a bimodal effect on TRPA1, producing current inhibition at millimolar concentrations. Second, it directly and stably activates mouse and human recombinant TRPV1, as well as TRPV1 channels in mouse sensory neurons. Finally, physiological temperatures enhance MO-induced TRPV1 stimulation. Our results refute the dogma that TRPA1 is the sole nocisensor for MO and motivate a revision of the putative roles of these channels in models of MO-induced pain and inflammation. We propose that TRPV1 has a generalized role in the detection of irritant botanical defensive traits and in the coevolution of multiple mammalian and plant species.


Assuntos
Mostardeira/toxicidade , Óleos de Plantas/toxicidade , Canais de Cátion TRPV/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cistite/induzido quimicamente , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Knockout , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Dor/induzido quimicamente , Dor/genética , Dor/metabolismo , Canal de Cátion TRPA1 , Canais de Cátion TRPV/genética , Fatores de Tempo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
13.
J Neural Transm (Vienna) ; 118(5): 699-712, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21210284

RESUMO

The deposition of amyloid-ß protein (Aß) in the brain is a hallmark of Alzheimer's disease (AD). Apolipoprotein E (apoE) is involved in the clearance of Aß from brain and the APOE ε4 allele is a major risk factor for sporadic AD. We have recently shown that apoE is drained into the perivascular space (PVS), where it co-localizes with Aß. To further clarify the role of apoE in perivascular clearance of Aß, we studied apoE-transgenic mice over-expressing human apoE4 either in astrocytes (GE4) or in neurons (TE4). These animals were crossbred with amyloid precursor protein (APP)-transgenic mice and with APP-presenilin-1 (APP-PS1) double transgenic mice. Using an antibody that specifically detects human apoE (h-apoE), we observed that astroglial expression of h-apoE in GE4 mice leads to its perivascular drainage, whereas neuronal expression in TE4 mice does not, indicating that neuron-derived apoE is usually not the subject of perivascular drainage. However, h-apoE was observed not only in the PVS of APP-GE4 and APP-PS1-GE4 mice, but also in that of APP-TE4 and APP-PS1-TE4 mice. In all these mouse lines, we found co-localization of neuron-derived h-apoE and Aß in the PVS. Aß and h-apoE were also found in the cytoplasm of perivascular astrocytes indicating that astrocytes take up the neuron-derived apoE bound to Aß, presumably prior to its clearance into the PVS. The uptake of apoE-Aß complexes into glial cells was further investigated in glioblastoma cells. It was mediated by α(2)macroglobulin receptor/low density lipoprotein receptor-related protein (LRP-1) and inhibited by adding receptor-associated protein (RAP). It results in endosomal Aß accumulation within these cells. These results suggest that neuronal apoE-Aß complexes, but not neuronal apoE alone, are substrates for LRP-1-mediated astroglial uptake, transcytosis, and subsequent perivascular drainage. Thus, the production of Aß and its interaction with apoE lead to the pathological perivascular drainage of neuronal apoE and provide insight into the pathological interactions of Aß with neuronal apoE metabolism.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/patologia , Ventrículos Cerebrais/fisiopatologia , Neurônios/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Apolipoproteínas E/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Camundongos Transgênicos , Neuroblastoma/patologia , Neuroblastoma/fisiopatologia , Presenilina-1/genética , Receptores de LDL/metabolismo , Transfecção , Proteínas Supressoras de Tumor/metabolismo
14.
Biochim Biophys Acta ; 1802(10): 808-18, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20332023

RESUMO

Alzheimer's dementia is developing ever more as a complex syndrome with various unknown genetic and epigenetic contributions. These are compounded on and exacerbating the underlying amyloid and tau pathology that remain the basis of the pathological definition of Alzheimer's disease. Here, we present a selection of aspects of recent bigenic and virus-based mouse strains, developed as pre-clinical models for Alzheimer's disease. We discuss newer features in the context of the characteristics defined in previously validated transgenic models. We focus on specific aspects of single and multiple transgenic mouse models for Alzheimer's disease and for tauopathies, rather than providing an exhaustive list of all available models. We concentrate on the content of information related to neurodegeneration and disease mechanisms. We pay attention to aspects and defects that are predicted by the models and can be tested in humans. We discuss implications that help translate the fundamental knowledge into clinical, diagnostic and therapeutic applications. We elaborate on the increasing knowledge extracted from transgenic models and from newer adeno-associated viral models. We advocate this combination as a valuable strategy to study molecular, cellular and system-related pathogenic mechanisms in AD and tauopathies. We believe that innovative animal models remain needed to critically test current views, to identify and validate therapeutic targets, to allow testing of compounds, to help understand and eventually treat tauopathies, including Alzheimer's disease.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/virologia , Modelos Animais de Doenças , Tauopatias/genética , Tauopatias/virologia , Doença de Alzheimer/patologia , Animais , Humanos , Camundongos , Camundongos Transgênicos , Tauopatias/patologia
15.
PLoS One ; 4(10): e7280, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19794916

RESUMO

In Alzheimer's disease tauopathy is considered secondary to amyloid, and the duality obscures their relation and the definition of their respective contributions.Transgenic mouse models do not resolve this problem conclusively, i.e. the relative hierarchy of amyloid and tau pathology depends on the actual model and the genes expressed or inactivated. Here, we approached the problem in non-transgenic models by intracerebral injection of adeno-associated viral vectors to express protein tau or amyloid precursor protein in the hippocampus in vivo. AAV-APP mutant caused neuronal accumulation of amyloid peptides, and eventually amyloid plaques at 6 months post-injection, but with only marginal hippocampal cell-death. In contrast, AAV-Tau, either wild-type or mutant P301L, provoked dramatic degeneration of pyramidal neurons in CA1/2 and cortex within weeks. Tau-mediated neurodegeneration proceeded without formation of large fibrillar tau-aggregates or tangles, but with increased expression of cell-cycle markers.We present novel AAV-based models, which demonstrate that protein tau mediates pyramidal neurodegeneration in vivo. The data firmly support the unifying hypothesis that post-mitotic neurons are forced to re-enter the cell-cycle in primary and secondary tauopathies, including Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Dependovirus/metabolismo , Doenças Neurodegenerativas/metabolismo , Emaranhados Neurofibrilares/metabolismo , Proteínas tau/química , Animais , Ciclo Celular , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Estrutura Terciária de Proteína
16.
Biochim Biophys Acta ; 1783(7): 1381-95, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18298958

RESUMO

Budding yeast Saccharomyces cerevisiae has proven to be a valuable model organism for studying fundamental cellular processes across the eukaryotic kingdom including man. In this respect, complementation assays, in which the yeast protein is replaced by a homologous protein from another organism, have been very instructive. A newer trend is to use the yeast cell factory as a toolbox to understand cellular processes controlled by proteins for which the yeast lacks functional counterparts. An increasing number of studies have indicated that S. cerevisiae is a suitable model system to decipher molecular mechanisms involved in a variety of neurodegenerative disorders caused by aberrant protein folding. Here we review the current knowledge gained by the use of so-called humanized yeasts in the field of Huntington's, Parkinson's and Alzheimer's diseases.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Doença de Huntington/metabolismo , Degeneração Neural/metabolismo , Doença de Parkinson/metabolismo , Saccharomyces cerevisiae/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Apoptose/fisiologia , Proteínas de Choque Térmico/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/fisiopatologia , Modelos Biológicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/fisiopatologia , Peptídeos/metabolismo , Dobramento de Proteína , Leveduras , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo
17.
J Clin Invest ; 117(11): 3453-62, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17948126

RESUMO

Here we provide evidence for a critical role of the transient receptor potential cation channel, subfamily V, member 4 (TRPV4) in normal bladder function. Immunofluorescence demonstrated TRPV4 expression in mouse and rat urothelium and vascular endothelium, but not in other cell types of the bladder. Intracellular Ca2+ measurements on urothelial cells isolated from mice revealed a TRPV4-dependent response to the selective TRPV4 agonist 4alpha-phorbol 12,13-didecanoate and to hypotonic cell swelling. Behavioral studies demonstrated that TRPV4-/- mice manifest an incontinent phenotype but show normal exploratory activity and anxiety-related behavior. Cystometric experiments revealed that TRPV4-/- mice exhibit a lower frequency of voiding contractions as well as a higher frequency of nonvoiding contractions. Additionally, the amplitude of the spontaneous contractions in explanted bladder strips from TRPV4-/- mice was significantly reduced. Finally, a decreased intravesical stretch-evoked ATP release was found in isolated whole bladders from TRPV4-/- mice. These data demonstrate a previously unrecognized role for TRPV4 in voiding behavior, raising the possibility that TRPV4 plays a critical role in urothelium-mediated transduction of intravesical mechanical pressure.


Assuntos
Canais de Cátion TRPV/metabolismo , Bexiga Urinária/metabolismo , Micção/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Comportamento Animal/fisiologia , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Ratos , Canais de Cátion TRPV/genética , Bexiga Urinária/anatomia & histologia , Urodinâmica , Urotélio/citologia , Urotélio/metabolismo
18.
Neurobiol Dis ; 19(3): 386-99, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16023581

RESUMO

A complex mutation in DFNA5, resulting in exon 8 skipping, causes autosomal dominant hearing impairment, which starts in the high frequencies between 5 and 15 years of age and progressively affects all frequencies. To study its function in vivo, Dfna5 knockout mice were generated through the deletion of exon 8, simultaneously mimicking the human mutation. To test the hearing impairment, frequency-specific Auditory Brainstem Response (ABR) measurements were performed at different ages in two genetic backgrounds (C57Bl/6J and CBA/Ca), but no differences between Dfna5-/- and Dfna5+/+ mice could be demonstrated. Morphological studies demonstrated significant differences in the number of fourth row outer hair cells between Dfna5-/- mice and their wild-type littermates. These results were obtained in both genetic backgrounds, albeit with opposite effects. In contrast to the results obtained in Dfna5-/- zebrafish, we did not observe different UDP-glucose dehydrogenase and hyaluronic acid levels in Dfna5-/- mice when compared to Dfna5+/+ mice.


Assuntos
Cóclea/ultraestrutura , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Células Ciliadas Auditivas Externas/ultraestrutura , Receptores de Estrogênio/deficiência , Animais , Western Blotting , Modelos Animais de Doenças , Genótipo , Perda Auditiva/congênito , Perda Auditiva/patologia , Ácido Hialurônico/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Uridina Difosfato Glucose Desidrogenase/metabolismo
19.
Brain ; 128(Pt 6): 1442-53, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15817521

RESUMO

Neuritic plaques in the brain of Alzheimer's disease patients are characterized by beta-amyloid deposits associated with a glia-mediated inflammatory response. Non-steroidal anti-inflammatory drug (NSAID) therapy reduces Alzheimer's disease risk and ameliorates microglial reactivity in Alzheimer's disease brains; however, the molecular mechanisms subserving this effect are not yet clear. Since several NSAIDs bind to and activate the nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARgamma) which acts to inhibit the expression of proinflammatory genes, this receptor appears a good candidate to mediate the observed anti-inflammatory effects. Recent data in vitro suggested that NSAIDs negatively regulate microglial activation and immunostimulated amyloid precursor protein processing via PPARgamma activation. We report that an acute 7 day oral treatment of 10-month-old APPV717I mice with the PPARgamma agonist pioglitazone or the NSAID ibuprofen resulted in a reduction in the number of activated microglia and reactive astrocytes in the hippocampus and cortex. Drug treatment reduced the expression of the proinflammatory enzymes cyclooxygenase 2 (COX2) and inducible nitric oxide synthase (iNOS). In parallel to the suppression of inflammatory markers, pioglitazone and ibuprofen treatment decreased beta-secretase-1 (BACE1) mRNA and protein levels. Importantly, we observed a significant reduction of the total area and staining intensity of Abeta1-42-positive amyloid deposits in the hippocampus and cortex. Additionally, animals treated with pioglitazone revealed a 27% reduction in the levels of soluble Abeta1-42 peptide. These findings demonstrate that anti-inflammatory drugs can act rapidly to inhibit inflammatory responses in the brain and negatively modulate amyloidogenesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Ibuprofeno/uso terapêutico , Microglia/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Tiazolidinedionas/uso terapêutico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide , Amiloidose/tratamento farmacológico , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Ciclo-Oxigenase 2 , Modelos Animais de Doenças , Endopeptidases , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Técnicas Imunoenzimáticas , Camundongos , Camundongos Transgênicos , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , PPAR gama/agonistas , Pioglitazona , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Mensageiro/genética
20.
Am J Pathol ; 165(5): 1621-31, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15509532

RESUMO

The generation of amyloid peptides (Abeta) from the amyloid precursor protein (APP) is initiated by beta-secretase (BACE), whereas subsequent gamma-secretase cleavage mediated by presenilin-1, produces Abeta peptides mainly of 40 or 42 amino acids long. In addition, alternative beta'-cleavage of APP at position 11 of the amyloid sequence results in N-truncated Abeta(11-40/42) peptides, but the functional significance or pathological impact is unknown. Here we demonstrate that in the brain of BACE x APP[V717I] double-transgenic mice, amyloidogenic processing at both Asp1 and Glu11 is increased resulting in more and different Abeta species and APP C-terminal fragments. Pathologically, BACE significantly increased the number of diffuse and senile amyloid plaques in old double-transgenic mice. Unexpectedly, vascular amyloid deposition was dramatically lower in the same BACE x APP[V717I] double-transgenic mice, relative to sex- and age-matched APP[V717I] single-transgenic mice in the same genetic background. The tight inverse relation of vascular amyloid to the levels of the less soluble N-terminally truncated Abeta peptides is consistent with the hypothesis that vascular amyloid deposition depends on drainage of excess tissue Abeta. This provides biochemical evidence in vivo for the preferential contribution of N-truncated Abeta to parenchymal amyloid deposition in contrast to vascular amyloid pathology.


Assuntos
Amiloide/metabolismo , Encéfalo/metabolismo , Endopeptidases/fisiologia , Fatores Etários , Amiloide/química , Amiloide/genética , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Ácido Aspártico Endopeptidases , Encéfalo/patologia , Angiopatia Amiloide Cerebral/patologia , Córtex Cerebral/patologia , Endopeptidases/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Genótipo , Imuno-Histoquímica , Modelos Lineares , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Peptídeos/química , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA