Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(49): e2211999119, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36442130

RESUMO

Impairments in neural lysosomal- and autophagic-mediated degradation of cellular debris contribute to neuritic dystrophy and synaptic loss. While these are well-characterized features of neurodegenerative disorders such as Alzheimer's disease (AD), the upstream cellular processes driving deficits in pathogenic protein mishandling are less understood. Using a series of fluorescent biosensors and optical imaging in model cells, AD mouse models and human neurons derived from AD patients, we reveal a previously undescribed cellular signaling cascade underlying protein mishandling mediated by intracellular calcium dysregulation, an early component of AD pathogenesis. Increased Ca2+ release via the endoplasmic reticulum (ER)-resident ryanodine receptor (RyR) is associated with reduced expression of the lysosome proton pump vacuolar-ATPase (vATPase) subunits (V1B2 and V0a1), resulting in lysosome deacidification and disrupted proteolytic activity in AD mouse models and human-induced neurons (HiN). As a result of impaired lysosome digestive capacity, mature autophagosomes with hyperphosphorylated tau accumulated in AD murine neurons and AD HiN, exacerbating proteinopathy. Normalizing AD-associated aberrant RyR-Ca2+ signaling with the negative allosteric modulator, dantrolene (Ryanodex), restored vATPase levels, lysosomal acidification and proteolytic activity, and autophagic clearance of intracellular protein aggregates in AD neurons. These results highlight that prior to overt AD histopathology or cognitive deficits, aberrant upstream Ca2+ signaling disrupts lysosomal acidification and contributes to pathological accumulation of intracellular protein aggregates. Importantly, this is demonstrated in animal models of AD, and in human iPSC-derived neurons from AD patients. Furthermore, pharmacological suppression of RyR-Ca2+ release rescued proteolytic function, revealing a target for therapeutic intervention that has demonstrated effects in clinically-relevant assays.


Assuntos
Doença de Alzheimer , Cálcio , Humanos , Camundongos , Animais , Proteólise , Agregados Proteicos , Cálcio da Dieta , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Dantroleno , Lisossomos , Modelos Animais de Doenças
2.
Int J Mol Sci ; 21(3)2020 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-32033164

RESUMO

Traditional approaches to studying Alzheimer's disease (AD) using mouse models and cell lines have advanced our understanding of AD pathogenesis. However, with the growing divide between model systems and clinical therapeutic outcomes, the limitations of these approaches are increasingly apparent. Thus, to generate more clinically relevant systems that capture pathological cascades within human neurons, we generated human-induced neurons (HiNs) from AD and non-AD individuals to model cell autonomous disease properties. We selected an AD patient population expressing mutations in presenilin 1 (mPS1), which is linked to increased amyloid production, tau pathology, and calcium signaling abnormalities, among other features. While these AD components are detailed in model systems, they have yet to be collectively identified in human neurons. Thus, we conducted molecular, immune-based, electrophysiological, and calcium imaging studies to establish patterns of cellular pathology in this patient population. We found that mPS1 HiNs generate increased Aß42 and hyperphosphorylated tau species relative to non-AD controls, and exaggerated ER calcium responses that are normalized with ryanodine receptor (RyR) negative allosteric modulators. The inflammasome product, interleukin-18 (IL-18), also increased PS1 expression. This work highlights the potential for HiNs to model AD pathology and validates their role in defining cellular pathogenesis and their utility for therapeutic screening.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Mutação/genética , Neurônios/patologia , Presenilina-1/genética , Regulação Alostérica/fisiologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Retículo Endoplasmático/metabolismo , Humanos , Inflamassomos/genética , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Neurônios/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Proteínas tau/metabolismo
3.
J Neurosci ; 38(19): 4505-4520, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29650696

RESUMO

Neuropeptide Y (NPY) expression is tightly linked with the development of stress resilience in rodents and humans. Local NPY injections targeting the basolateral amygdala (BLA) produce long-term behavioral stress resilience in male rats via an unknown mechanism. Previously, we showed that activation of NPY Y1 receptors hyperpolarizes BLA principal neurons (PNs) through inhibition of the hyperpolarization-activated, depolarizing H-current, Ih The present studies tested whether NPY treatment induces stress resilience by modulating Ih NPY (10 pmol) was delivered daily for 5 d bilaterally into the BLA to induce resilience; thereafter, the electrophysiological properties of PNs and the expression of Ih in the BLA were characterized. As reported previously, increases in social interaction (SI) times persisted weeks after completion of NPY administration. In vitro intracellular recordings showed that repeated intra-BLA NPY injections resulted in hyperpolarization of BLA PNs at 2 weeks (2W) and 4 weeks (4W) after NPY treatment. At 2W, spontaneous IPSC frequencies were increased, whereas at 4W, resting Ih was markedly reduced and accompanied by decreased levels of HCN1 mRNA and protein expression in BLA. Knock-down of HCN1 channels in the BLA with targeted delivery of lentivirus containing HCN1-shRNA increased SI beginning 2W after injection and induced stress resilience. NPY treatment induced sequential, complementary changes in the inputs to BLA PNs and their postsynaptic properties that reduce excitability, a mechanism that contributes to less anxious behavior. Furthermore, HCN1 knock-down mimicked the increases in SI and stress resilience observed with NPY, indicating the importance of Ih in stress-related behavior.SIGNIFICANCE STATEMENT Resilience improves mental health outcomes in response to adverse situations. Neuropeptide Y (NPY) is associated with decreased stress responses and the expression of resilience in rodents and humans. Single or repeated injections of NPY into the basolateral amygdala (BLA) buffer negative behavioral effects of stress and induce resilience in rats, respectively. Here, we demonstrate that repeated administration of NPY into the BLA unfolds several cellular mechanisms that decrease the activity of pyramidal output neurons. One key mechanism is a reduction in levels of the excitatory ion channel HCN1. Moreover, shRNA knock-down of HCN1 expression in BLA recapitulates some of the actions of NPY and causes potent resilience to stress, indicating that this channel may be a possible target for therapy.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Canais de Potássio/efeitos dos fármacos , Resiliência Psicológica/efeitos dos fármacos , Tonsila do Cerebelo/citologia , Animais , Ansiedade/genética , Ansiedade/psicologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Relações Interpessoais , Masculino , Microinjeções , Neuropeptídeo Y/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
4.
Mol Ther ; 26(6): 1539-1551, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29628304

RESUMO

Alterations in amyloid beta precursor protein (APP) have been implicated in cognitive decline in Alzheimer's disease (AD), which is accelerated in Down syndrome/Trisomy 21 (DS/TS21), likely due to the extra copy of the APP gene, located on chromosome 21. Proteolytic cleavage of APP generates amyloid-ß (Aß) peptide, the primary component of senile plaques associated with AD. Reducing Aß production is predicted to lower plaque burden and mitigate AD symptoms. Here, we designed a splice-switching antisense oligonucleotide (SSO) that causes skipping of the APP exon that encodes proteolytic cleavage sites required for Aß peptide production. The SSO induced exon skipping in Down syndrome cell lines, resulting in a reduction of Aß. Treatment of mice with the SSO resulted in widespread distribution in the brain accompanied by APP exon skipping and a reduction of Aß. Overall, we show that an alternatively spliced isoform of APP encodes a cleavage-incompetent protein that does not produce Aß peptide and that promoting the production of this isoform with an SSO can reduce Aß in vivo. These findings demonstrate the utility of using SSOs to induce a spliced isoform of APP to reduce Aß as a potential approach for treating AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Oligonucleotídeos Antissenso/genética , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Síndrome de Down/genética , Síndrome de Down/metabolismo , Éxons/genética , Camundongos
5.
Stem Cells ; 30(3): 500-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22162276

RESUMO

The generation of new neurons in the mammalian hippocampus continues throughout life, and lineage progression is regulated by transcription factors, local cues, and environmental influences. The ability to direct stem/progenitor cell fate in situ may have therapeutic potential. Using an in vivo retroviral delivery and lineage tracing approach, we compare the lineage-instruction factors, Pax6 and Olig2, and demonstrate that both participate in regulation of adult hippocampal neurogenesis in adult rats. We show that overexpression of the proneuronal factor Pax6 pushes neuronal precursor cells to early maturation and increases the frequency of neuronal phenotypes. However, Pax6 overexpression results in no net increase in neurogenesis at 3 weeks. Blocking of Olig2 function reduces and slows neuronal commitment and differentiation and decreases net neurogenesis. Altering expression of both factors also changes gliogenesis. Our results establish that Pax6 decreases the number of Neuron-Glia 2 progenitor cells and prevents oligodendrocytic lineage commitment, while repression of Olig2 results in an expanded astrocytic lineage. We conclude that selectively modifying transcriptional cues within hippocampal progenitor cells is sufficient to induce a cell fate switch, thus altering the neurogenesis-gliogenesis ratio. In addition, our data show the competence of multiple progenitor lineages to respond divergently to the same signal. Therefore, directing instructive cues to select phenotype and developmental stage could be critical to achieve precise outcomes in cell genesis. Further understanding the regulation of lineage progression in all progenitor populations within the target region will be important for developing therapeutic strategies to direct cell fate for brain repair.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas do Olho/metabolismo , Hipocampo/citologia , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neuroglia/citologia , Neurônios/citologia , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem da Célula , Proliferação de Células , Forma Celular , Sobrevivência Celular , Proteínas do Olho/genética , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fenótipo , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Medicina Regenerativa , Proteínas Repressoras/genética , Transdução de Sinais
6.
Front Neurosci ; 17: 1237176, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37662111

RESUMO

Adult tissue stem cells contribute to tissue homeostasis and repair but the long-lived neurons in the human adult cerebral cortex are not replaced, despite evidence for a limited regenerative response. However, the adult cortex contains a population of proliferating oligodendrocyte progenitor cells (OPCs). We examined the capacity of rat cortical OPCs to be re-specified to a neuronal lineage both in vitro and in vivo. Expressing the developmental transcription factor Neurogenin2 (Ngn2) in OPCs isolated from adult rat cortex resulted in their expression of early neuronal lineage markers and genes while downregulating expression of OPC markers and genes. Ngn2 induced progression through a neuronal lineage to express mature neuronal markers and functional activity as glutamatergic neurons. In vivo retroviral gene delivery of Ngn2 to naive adult rat cortex ensured restricted targeting to proliferating OPCs. Ngn2 expression in OPCs resulted in their lineage re-specification and transition through an immature neuronal morphology into mature pyramidal cortical neurons with spiny dendrites, axons, synaptic contacts, and subtype specification matching local cytoarchitecture. Lineage re-specification of rat cortical OPCs occurred without prior injury, demonstrating these glial progenitor cells need not be put into a reactive state to achieve lineage reprogramming. These results show it may be feasible to precisely engineer additional neurons directly in adult cerebral cortex for experimental study or potentially for therapeutic use to modify dysfunctional or damaged circuitry.

7.
Am J Pathol ; 178(1): 306-12, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21224067

RESUMO

Proteases that degrade the amyloid-ß peptide (Aß) are important in protecting against Alzheimer's disease (AD), and understanding these proteases is critical to understanding AD pathology. Endopeptidases sensitive to inhibition by thiorphan and phosphoramidon are especially important, because these inhibitors induce dramatic Aß accumulation (∼30- to 50-fold) and pathological deposition in rodents. The Aß-degrading enzyme neprilysin (NEP) is the best known target of these inhibitors. However, genetic ablation of NEP results in only modest increases (∼1.5- to 2-fold) in Aß, indicating that other thiorphan/phosphoramidon-sensitive endopeptidases are at work. Of particular interest is the NEP homolog neprilysin 2 (NEP2), which is thiorphan/phosphoramidon-sensitive and degrades Aß. We investigated the role of NEP2 in Aß degradation in vivo through the use of gene knockout and transgenic mice. Mice deficient for the NEP2 gene showed significant elevations in total Aß species in the hippocampus and brainstem/diencephalon (∼1.5-fold). Increases in Aß accumulation were more dramatic in NEP2 knockout mice crossbred with APP transgenic mice. In NEP/NEP2 double-knockout mice, Aß levels were marginally increased (∼1.5- to 2-fold), compared with NEP(-/-)/NEP2(+/+) controls. Treatment of these double-knockout mice with phosphoramidon resulted in elevations of Aß, suggesting that yet other NEP-like Aß-degrading endopeptidases are contributing to Aß catabolism.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Neprilisina/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Glicopeptídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neprilisina/genética
8.
Nature ; 438(7068): 662-6, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16244648

RESUMO

Anxiety and fear are normal emotional responses to threatening situations. In human anxiety disorders--such as panic disorder, obsessive-compulsive disorder, post-traumatic stress disorder, social phobia, specific phobias and generalized anxiety disorder--these responses are exaggerated. The molecular mechanisms involved in the regulation of normal and pathological anxiety are mostly unknown. However, the availability of different inbred strains of mice offers an excellent model system in which to study the genetics of certain behavioural phenotypes. Here we report, using a combination of behavioural analysis of six inbred mouse strains with quantitative gene expression profiling of several brain regions, the identification of 17 genes with expression patterns that correlate with anxiety-like behavioural phenotypes. To determine if two of the genes, glyoxalase 1 and glutathione reductase 1, have a causal role in the genesis of anxiety, we performed genetic manipulation using lentivirus-mediated gene transfer. Local overexpression of these genes in the mouse brain resulted in increased anxiety-like behaviour, while local inhibition of glyoxalase 1 expression by RNA interference decreased the anxiety-like behaviour. Both of these genes are involved in oxidative stress metabolism, linking this pathway with anxiety-related behaviour.


Assuntos
Ansiedade/enzimologia , Ansiedade/genética , Encéfalo/metabolismo , Perfilação da Expressão Gênica , Glutationa Redutase/metabolismo , Lactoilglutationa Liase/metabolismo , Animais , Ansiedade/fisiopatologia , Feminino , Regulação da Expressão Gênica , Glutationa Redutase/genética , Lactoilglutationa Liase/genética , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos , Estresse Oxidativo/genética , Fenótipo , Transdução Genética
9.
Front Bioeng Biotechnol ; 9: 658498, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777923

RESUMO

Direct cellular reprogramming exhibits distinct advantages over reprogramming from an induced pluripotent stem cell intermediate. These include a reduced risk of tumorigenesis and the likely preservation of epigenetic data. In vitro direct reprogramming approaches primarily aim to model the pathophysiological development of neurological disease and identify therapeutic targets, while in vivo direct reprogramming aims to develop treatments for various neurological disorders, including cerebral injury and cancer. In both approaches, there is progress toward developing increased control of subtype-specific production of induced neurons. A majority of research primarily utilizes fibroblasts as the donor cells. However, there are a variety of other somatic cell types that have demonstrated the potential for reprogramming into induced neurons. This review highlights studies that utilize non-fibroblastic cell sources for reprogramming, such as astrocytes, olfactory ensheathing cells, peripheral blood cells, Müller glia, and more. We will examine benefits and obstructions for translation into therapeutics or disease modeling, as well as efficiency of the conversion. A summary of donor cells, induced neuron types, and methods of induction is also provided.

10.
J Neurotrauma ; 38(5): 665-676, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33176547

RESUMO

Traumatic brain injury (TBI) increases the risk for dementias including Alzheimer's disease (AD) and chronic traumatic encephalopathy. Further, both human and animal model data indicate that amyloid-beta (Aß) peptide accumulation and its production machinery are upregulated by TBI. Considering the clear link between chronic Aß elevation and AD as well as tau pathology, the role(s) of Aß in TBI is of high importance. Endopeptidases, including the neprilysin (NEP)-like enzymes, are key mediators of Aß clearance and may affect susceptibility to pathology post-TBI. Here, we use a "humanized" mouse model of Aß production, which expresses normal human amyloid-beta precursor protein (APP) under its natural transcriptional regulation and exposed them to a more clinically relevant repeated closed-head TBI paradigm. These transgenic mice also were crossed with mice deficient for the Aß degrading enzymes NEP or NEP2 to assess models of reduced cerebral Aß clearance in our TBI model. Our results show that the presence of the human form of Aß did not exacerbate motor (Rotarod) and spatial learning/memory deficits (Morris water maze) post-injuries, while potentially reduced anxiety (Open Field) was observed. NEP and NEP2 deficiency also did not exacerbate these deficits post-injuries and was associated with protection from motor (NEP and NEP2) and spatial learning/memory deficits (NEP only). These data suggest that normally regulated expression of wild-type human APP/Aß does not contribute to deficits acutely after TBI and may be protective at this stage of injury.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Comportamento Animal/fisiologia , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/psicologia , Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/psicologia , Animais , Lesões Encefálicas Traumáticas/complicações , Modelos Animais de Doenças , Traumatismos Cranianos Fechados/complicações , Humanos , Aprendizagem em Labirinto/fisiologia , Camundongos Transgênicos , Teste de Desempenho do Rota-Rod
11.
J Neurosci ; 29(4): 1115-25, 2009 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-19176820

RESUMO

The endopeptidase neprilysin (NEP) is a major amyloid-beta (Abeta) degrading enzyme and has been implicated in the pathogenesis of Alzheimer's disease. Because NEP cleaves substrates other than Abeta, we investigated the potential role of NEP-mediated processing of neuropeptides in the mechanisms of neuroprotection in vivo. Overexpression of NEP at low levels in transgenic (tg) mice affected primarily the levels of neuropeptide Y (NPY) compared with other neuropeptides. Ex vivo and in vivo studies in tg mice and in mice that received lentiviral vector injections showed that NEP cleaved NPY into C-terminal fragments (CTFs), whereas silencing NEP reduced NPY processing. Immunoblot and mass spectrometry analysis showed that NPY 21-36 and 31-36 were the most abundant fragments generated by NEP activity in vivo. Infusion of these NPY CTFs into the brains of APP (amyloid precursor protein) tg mice ameliorated the neurodegenerative pathology in this model. Moreover, the amidated NPY CTFs protected human neuronal cultures from the neurotoxic effects of Abeta. This study supports the possibility that the NPY CTFs generated during NEP-mediated proteolysis might exert neuroprotective effects in vivo. This function of NEP represents a unique example of a proteolytic enzyme with dual action, namely, degradation of Abeta as well as processing of NPY.


Assuntos
Neprilisina/química , Neprilisina/metabolismo , Degeneração Neural/prevenção & controle , Neuropeptídeo Y/uso terapêutico , Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Benzazepinas/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/genética , Vetores Genéticos/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Neprilisina/genética , Degeneração Neural/etiologia , Fatores de Crescimento Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeo Y/química , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/uso terapêutico , Receptores de Neuropeptídeo Y/antagonistas & inibidores
12.
Nat Neurosci ; 8(10): 1343-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16136043

RESUMO

In Alzheimer disease, increased beta-secretase (BACE1) activity has been associated with neurodegeneration and accumulation of amyloid precursor protein (APP) products. Thus, inactivation of BACE1 could be important in the treatment of Alzheimer disease. In this study, we found that lowering BACE1 levels using lentiviral vectors expressing siRNAs targeting BACE1 reduced amyloid production and the neurodegenerative and behavioral deficits in APP transgenic mice, a model of Alzheimer disease. Our results suggest that lentiviral vector delivery of BACE1 siRNA can specifically reduce the cleavage of APP and neurodegeneration in vivo and indicate that this approach could have potential therapeutic value for treatment of Alzheimer disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , RNA Interferente Pequeno/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Ácido Aspártico Endopeptidases , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Clonagem Molecular , Modelos Animais de Doenças , Endopeptidases/química , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Vetores Genéticos/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imuno-Histoquímica/métodos , Lentivirus/fisiologia , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Dados de Sequência Molecular , Comportamento Espacial/efeitos dos fármacos , Fatores de Tempo
13.
BMC Neurosci ; 9: 109, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19014502

RESUMO

BACKGROUND: Proteolytic degradation has emerged as a key pathway involved in controlling levels of the Alzheimer's disease (AD)-associated amyloid-beta (Abeta) peptide in the brain. The endopeptidase, neprilysin, has been implicated as a major Abeta degrading enzyme in mice and humans. Previous short and intermediate term studies have shown the potential therapeutic application of neprilysin by delivering this enzyme into the brain of APP transgenic mice using gene transfer with viral vectors. However the effects of long-term neprilysin gene transfer on other aspects of Abeta associated pathology have not been explored yet in APP transgenic mice. RESULTS: We show that the sustained expression of neprilysin for up to 6 months lowered not only the amyloid plaque load but also reduced the levels of intracellular Abeta immunoreactivity. This was associated with improved behavioral performance in the water maze and ameliorated the dendritic and synaptic pathology in the APP transgenic mice. CONCLUSION: These data support the possibility that long-term neprilysin gene therapy improves behavioral and neurodegenerative pathology by reducing intracellular Abeta.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Encéfalo/metabolismo , Neprilisina/genética , Doença de Alzheimer/patologia , Animais , Comportamento Animal , Encéfalo/patologia , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Líquido Intracelular/metabolismo , Lentivirus/genética , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Neprilisina/biossíntese , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/genética , Placa Amiloide/patologia , Tempo
14.
EMBO Mol Med ; 8(4): 328-45, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26902204

RESUMO

Apolipoprotein E receptor 2 (ApoER2) is an apolipoprotein E receptor involved in long-term potentiation, learning, and memory. Given its role in cognition and its association with the Alzheimer's disease (AD) risk gene, apoE, ApoER2 has been proposed to be involved in AD, though a role for the receptor in the disease is not clear. ApoER2 signaling requires amino acids encoded by alternatively spliced exon 19. Here, we report that the balance of ApoER2 exon 19 splicing is deregulated in postmortem brain tissue from AD patients and in a transgenic mouse model of AD To test the role of deregulated ApoER2 splicing in AD, we designed an antisense oligonucleotide (ASO) that increases exon 19 splicing. Treatment of AD mice with a single dose of ASO corrected ApoER2 splicing for up to 6 months and improved synaptic function and learning and memory. These results reveal an association between ApoER2 isoform expression and AD, and provide preclinical evidence for the utility of ASOs as a therapeutic approach to mitigate Alzheimer's disease symptoms by improving ApoER2 exon 19 splicing.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Proteínas Relacionadas a Receptor de LDL/metabolismo , Oligonucleotídeos Antissenso/uso terapêutico , Splicing de RNA , Doença de Alzheimer/patologia , Animais , Encéfalo/fisiologia , Modelos Animais de Doenças , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Aprendizagem , Memória , Camundongos , Camundongos Transgênicos , Oligonucleotídeos Antissenso/genética , Resultado do Tratamento
15.
J Neurosci ; 23(6): 1992-6, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12657655

RESUMO

The degenerative process of Alzheimer's disease is linked to a shift in the balance between amyloid-beta (Abeta) production, clearance, and degradation. Neprilysin has recently been implicated as a major extracellular Abeta degrading enzyme in the brain. However, there has been no direct demonstration that neprilysin antagonizes the deposition of amyloid-beta in vivo. To address this issue, a lentiviral vector expressing human neprilysin (Lenti-Nep) was tested in transgenic mouse models of amyloidosis. We show that unilateral intracerebral injection of Lenti-Nep reduced amyloid-beta deposits by half relative to the untreated side. Furthermore, Lenti-Nep ameliorated neurodegenerative alterations in the frontal cortex and hippocampus of these transgenic mice. These data further support a role for neprilysin in regulating cerebral amyloid deposition and suggest that gene transfer approaches might have potential for the development of alternative therapies for Alzheimer's disease.


Assuntos
Amiloidose/genética , Amiloidose/terapia , Terapia Genética/métodos , Neprilisina/genética , Neprilisina/farmacologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Técnicas de Transferência de Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Rim/citologia , Rim/metabolismo , Lentivirus/genética , Camundongos , Camundongos Transgênicos , Neprilisina/biossíntese , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/efeitos dos fármacos , Placa Amiloide/patologia , Células-Tronco/metabolismo , Resultado do Tratamento
16.
J Mol Neurosci ; 22(1-2): 5-11, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14742905

RESUMO

That neprilysin (NEP) is a major Abeta peptide-degrading enzyme in vivo is shown by higher Abeta peptide levels in the brain of an NEP knockout mouse. In addition, we show that infusion of an NEPinhibitor, but not inhibitors of other peptidases, into the brains of an APP transgenic mouse elevates Abeta levels. We have investigated the use of NEP as a potential therapeutic agent to prevent the accumulation of Abeta peptides in the brain. Lentivirus expressing NEP was initially used to demonstrate the ability of the enzyme to reduce Abeta levels in a model CHO cell line and to make primary hippocampal neurons resistant to Abeta-mediated neurotoxicity. Injection of NEPexpressing lentivirus, but not inactive NEP-expressing lentivirus, GFP-expressing lentivirus, or vehicle, into the hippocampus of 12-20-mo-old hAPP transgenic mice led to an approx 50% reduction in the number of amyloid plaques. These studies provide the impetus for further investigating of the use of NEP in a gene transfer therapy paradigm to prevent the accumulation of Abeta and prevent or delay the onset of Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Regulação para Baixo/genética , Neprilisina/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Células CHO , Células Cultivadas , Cricetinae , Modelos Animais de Doenças , Resistência a Medicamentos/genética , Inibidores Enzimáticos/farmacologia , Feto , Terapia Genética/métodos , Vetores Genéticos , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Lentivirus/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neprilisina/antagonistas & inibidores , Neprilisina/genética , Placa Amiloide/genética , Placa Amiloide/metabolismo
17.
Front Aging Neurosci ; 6: 187, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25165447

RESUMO

Accumulation of the amyloid-beta (Aß) peptide is a central factor in Alzheimer's disease (AD) pathogenesis as supported by continuing evidence. This review concisely summarizes this evidence supporting a critical role for Aß in AD before discussing the clearance of this peptide. Mechanisms of clearance of Aß are critical for preventing pathological elevations in Aß concentration. Direct degradation of Aß by endopeptidases has emerged as one important pathway for clearance. Of particular interest are endopeptidases that are sensitive to the neprilysin (NEP) inhibitors thiorphan and phosphoramidon (i.e., are "NEP-like") as these inhibitors induce a dramatic increase in Aß levels in rodents. This review will focus on neprilysin-2 (NEP2), a NEP-like endopeptidase which cooperates with NEP to control Aß levels in the brain. The evidence for the involvement of NEP2 in AD is discussed as well as the therapeutic relevance with regards to gene therapy and the development of molecular markers for the disease.

18.
Front Aging Neurosci ; 6: 265, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25346686

RESUMO

Currently, there are very limited pharmaceutical interventions for Alzheimer's disease (AD) to alleviate the amyloid burden implicated in the pathophysiology of the disease. Alzheimer's disease is characterized immunohistologically by the accumulation of senile plaques in the brain with afflicted patients progressively losing short-term memory and, ultimately, cognition. Although significant improvements in clinical diagnosis and care for AD patients have been made, effective treatments for this devastating disease remain elusive. A key component of the amyloid burden of AD comes from accumulation of the amyloid-beta (Aß) peptide which comes from processing of the amyloid precursor protein (APP) by enzymes termed secretases, leading to production of these toxic Aß peptides of 40-42 amino acids. New therapeutic approaches for reducing Aß are warranted after the most logical avenues of inhibiting secretase activity appear less than optimal in ameliorating the progression of AD.Novel therapeutics may be gleaned from proteomics biomarker initiatives to yield detailed molecular interactions of enzymes and their potential substrates. Explicating the APPome by deciphering protein complexes forming in cells is a complementary approach to unveil novel molecular interactions with the amyloidogenic peptide precursor to both understand the biology and develop potential upstream drug targets. Utilizing these strategies we have identified EC 3.4.24.15 (EP24.15), a zinc metalloprotease related to neprilysin (NEP), with the ability to catabolize Aß 1-42 by examining first potential in silico docking and then verification by mass spectrometry. In addition, a hormone carrier protein, transthyreitin (TTR), was identified and with its abundance in cerebrospinal fluid (CSF), found to clear Aß by inhibiting formation of oligomeric forms of Aß peptide. The confluence of complementary strategies may allow new therapeutic avenues as well as biomarkers for AD that will aid in diagnosis, prognosis and treatment.

19.
Front Aging Neurosci ; 5: 43, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23986699

RESUMO

Neural stem cells are maintained in the subgranular layer of the dentate gyrus and in the subventricular zone in the adult mammalian brain throughout life. Neurogenesis is continuous, but its extent is tightly regulated by environmental factors, behavior, hormonal state, age, and brain health. Increasing evidence supports a role for new neurons in cognitive function in rodents. Recent evidence delineates significant similarities and differences between adult neurogenesis in rodents and humans. Being context-dependent, neurogenesis in the human brain might be manifested differently than in the rodent brain. Decline in neurogenesis may play a role in cognitive deterioration, leading to the development of progressive learning and memory disorders, such as Alzheimer's disease. This review discusses the different observations concerning neurogenesis in the rodent and human brain, and their functional implications for the healthy and diseased brain.

20.
Neurobiol Aging ; 33(5): 1001.e1-6, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21531043

RESUMO

Intracellular Ca(2+) dysregulation is an underlying component of Alzheimer's disease (AD) pathophysiology, and recent evidence implicates the ryanodine receptor (RyR) in the disease pathway. Three genes code for different RyR isoforms and each gene transcript gives rise to several alternatively spliced messenger RNAs (mRNAs). These variants confer distinct functionality to the RyR channel, such as altering Ca(2+) release properties or subcellular localization. Changes in RyR isoform expression and alternative splicing have not been examined for potential roles in AD pathogenesis. Here, we compare mRNA levels of the RyR2 and RyR3 isoforms as well as specific alternatively spliced variants across vulnerable brain regions from postmortem samples of individuals with no cognitive impairment (NCI), mild cognitive impairment (MCI), and AD. We find an increase in RyR2 transcripts in MCI brains compared with no cognitive impairment. In addition, there is a reduction in a RyR2 splice variant, associated with an antiapoptotic function, in MCI and AD brains. These alterations in RyR expression at early disease stages may reflect the onset of pathologic mechanisms leading to later neurodegeneration.


Assuntos
Doença de Alzheimer/metabolismo , Disfunção Cognitiva/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/biossíntese , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Feminino , Humanos , Masculino , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos adversos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA