Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
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.

2.
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
3.
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.

4.
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
5.
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
6.
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
7.
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
8.
Behav Brain Res ; 353: 242-249, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29490235

RESUMO

Aging and major depressive disorder are risk factors for dementia, including Alzheimer's Disease (AD), but the mechanism(s) linking depression and dementia are not known. Both AD and depression show greater prevalence in women. We began to investigate this connection using females of the genetic model of depression, the inbred Wistar Kyoto More Immobile (WMI) rat. These rats consistently display depression-like behavior compared to the genetically close control, the Wistar Kyoto Less Immobile (WLI) strain. Hippocampus-dependent contextual fear memory did not differ between young WLI and WMI females, but, by middle-age, female WMIs showed memory deficits compared to same age WLIs. This deficit, measured as duration of freezing in the fear provoking-context was not related to activity differences between the strains prior to fear conditioning. Hippocampal expression of AD-related genes, such as amyloid precursor protein, amyloid beta 42, beta secretase, synucleins, total and dephosphorylated tau, and synaptophysin, did not differ between WLIs and WMIs in either age group. However, hippocampal transcript levels of catalase (Cat) and hippocampal and frontal cortex expression of insulin-like growth factor 2 (Igf2) and Igf2 receptor (Igf2r) paralleled fear memory differences between middle-aged WLIs and WMIs. This data suggests that chronic depression-like behavior that is present in this genetic model is a risk factor for early spatial memory decline in females. The molecular mechanisms of this early memory decline likely involve the interaction of aging processes with the genetic components responsible for the depression-like behavior in this model.


Assuntos
Envelhecimento/metabolismo , Transtorno Depressivo/metabolismo , Hipocampo/metabolismo , Transtornos da Memória/metabolismo , Envelhecimento/psicologia , Tonsila do Cerebelo/metabolismo , Animais , Estudos de Coortes , Condicionamento Psicológico/fisiologia , Transtorno Depressivo/complicações , Transtorno Depressivo/genética , Modelos Animais de Doenças , Medo/fisiologia , Feminino , Lobo Frontal/metabolismo , Expressão Gênica , Predisposição Genética para Doença , Memória/fisiologia , Transtornos da Memória/complicações , Ratos Endogâmicos WKY , Especificidade da Espécie
9.
Artigo em Inglês | MEDLINE | ID: mdl-35517374

RESUMO

Introduction: Cognitive aids, such as a guideline for the management of severe local anaesthetic (LA) toxicity, are tools designed to help users complete a task. Human factors experts recommend the use of simulation to iteratively test and re-design these tools. The purpose of this study was to apply human factors engineering principles to the testing and iterative re-design of three existing cognitive aids used for the management of severe LA toxicity and to use these data to develop a 'new' cognitive aid. Methods: Twenty anaesthetist-anaesthetic assistant pairs were randomised into four groups. Each of the first three groups received one of three different existing cognitive aids during a standardised simulated LA toxicity crisis. Postsimulation semistructured interviews were conducted to identify features beneficial and detrimental to the format and usability of the aid. Synthesis of the interview data with established checklist design recommendations resulted in a prototype aid, which was subjected to further testing and re-design by the fourth group (five more pairs) under the same conditions thus creating the final iteration of the new aid. Results: Features of the new aid included a single-stream flowchart structure, single-sided, large-font design with colour contrast, simplified instructions and no need for calculations. This simplified tool contains only the information users reported as essential for the immediate crisis management. Conclusions: Utilisation of formative usability testing and simulation-based user-centred design resulted in a visually very different cognitive aid and reinforces the importance of designing aids in the context in which they are to be used. Simplified tools may be more appropriate for use in emergencies but more detailed guidelines may be necessary for training, education and development of local standard operating procedures. Iterative simulation-based testing and re-design is likely to be of assistance when developing aids for other crises, and to eliminate design failure as a confounder when investigating the relationship between use of cognitive aids and performance.

10.
J Neurotrauma ; 34(7): 1351-1363, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27762651

RESUMO

Repeat concussions (RC) can result in significant long-term neurological consequences and increased risk for neurodegenerative disease compared with single concussion (SC). Mechanisms underlying this difference are poorly understood and best elucidated using an animal model. To the best of our knowledge, there is no closed-head model in the adult rat using a commercially available device. We developed a novel and clinically relevant closed-head injury (CHI) model of both SC and RC in the adult rat using a controlled cortical impact (CCI) device. Adult rats received either a single or repeat CHI (three injuries, 48 h apart), and acute deficits in sensorimotor and locomotor function (foot fault; open field), memory (novel object), and anxiety (open field; corticosterone [CORT]) were measured. Assessment of cellular pathology was also conducted. Within the first week post-CHI, rats with SC or RC showed similar deficits in motor coordination, decreased locomotion, and higher resting CORT levels. Rats with an SC had memory deficits post-injury day (PID) 3 that recovered to sham levels by PID 7; however, rats with RC continued to show memory deficits. No obvious gross pathology was observed on the cortical surface or in coronal sections. Further examination showed thinning of the cortex and corpus callosum in RC animals compared with shams and increased axonal pathology in the corpus callosum of both SC and RC animals. Our data present a model of CHI that results in clinically relevant markers of concussion and an early differentiation between SC and RC.


Assuntos
Concussão Encefálica/fisiopatologia , Córtex Cerebral/patologia , Disfunção Cognitiva/fisiopatologia , Corpo Caloso/patologia , Modelos Animais de Doenças , Transtornos da Memória/fisiopatologia , Atividade Motora/fisiologia , Desempenho Psicomotor/fisiologia , Animais , Comportamento Animal/fisiologia , Concussão Encefálica/complicações , Concussão Encefálica/etiologia , Concussão Encefálica/patologia , Disfunção Cognitiva/etiologia , Masculino , Transtornos da Memória/etiologia , Ratos , Ratos Long-Evans
11.
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
12.
PLoS One ; 10(6): e0131266, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098332

RESUMO

Presenilin-1 (PS1), the catalytic core of the aspartyl protease γ-secretase, regulates adult neurogenesis. However, it is not clear whether the role of neurogenesis in hippocampal learning and memory is PS1-dependent, or whether PS1 loss of function in adult hippocampal neurogenesis can cause learning and memory deficits. Here we show that downregulation of PS1 in hippocampal neural progenitor cells causes progressive deficits in pattern separation and novelty exploration. New granule neurons expressing reduced PS1 levels exhibit decreased dendritic branching and dendritic spines. Further, they exhibit reduced survival. Lastly, we show that PS1 effect on neurogenesis is mediated via ß-catenin phosphorylation and notch signaling. Together, these observations suggest that impairments in adult neurogenesis induce learning and memory deficits and may play a role in the cognitive deficits observed in Alzheimer's disease.


Assuntos
Hipocampo/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Neurogênese/fisiologia , Presenilina-1/fisiologia , Animais , Giro Denteado/fisiologia , Aprendizagem por Discriminação/fisiologia , Regulação para Baixo , Técnicas de Silenciamento de Genes , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/fisiologia , Fosforilação , Receptores Notch/fisiologia , Transdução de Sinais/fisiologia , beta Catenina/fisiologia
13.
14.
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.

15.
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.

16.
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.

19.
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
20.
J Alzheimers Dis ; 28(2): 433-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22008264

RESUMO

Neprilysin-2 (NEP2), a close homolog of neprilysin (NEP), degrades amyloid-ß (Aß) and serves an important role in clearing Aß in vivo. We measured NEP2 and NEP mRNA levels from non-impaired (NI), mild cognitive impaired (MCI), and clinical Alzheimer's disease (AD) subjects in the mid-temporal gyrus, mid-frontal gyrus, caudate, and cerebellum. NEP2 activity levels were also determined. Our results indicate that NEP2 and NEP mRNA expression is altered in MCI subjects relative to NI subjects in AD-susceptible regions. NEP2 enzymatic activity was lowered in association with MCI and AD and was positively associated with cognitive function, independent of diagnostic category. Our finding that NEP2 expression and activity are altered in MCI is significant as these changes may potentially serve as preclinical markers for AD and reduced NEP2 activity may be associated with the development of AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Disfunção Cognitiva/patologia , Neprilisina/metabolismo , Doença de Alzheimer/metabolismo , Análise de Variância , Encéfalo/patologia , Linhagem Celular Transformada , Disfunção Cognitiva/metabolismo , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Leucócitos Mononucleares/metabolismo , Modelos Lineares , Masculino , Neprilisina/genética , RNA Mensageiro/metabolismo , Fatores Sexuais , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...