Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35008983

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease with a multifactorial etiology. A multitarget treatment that modulates multifaceted biological functions might be more effective than a single-target approach. Here, the therapeutic efficacy of combination treatment using anti-Aß antibody NP106 and curcumin analog TML-6 versus monotherapy was investigated in an APP/PS1 mouse model of AD. Our data demonstrate that both combination treatment and monotherapy attenuated brain Aß and improved the nesting behavioral deficit to varying degrees. Importantly, the combination treatment group had the lowest Aß levels, and insoluble forms of Aß were reduced most effectively. The nesting performance of APP/PS1 mice receiving combination treatment was better than that of other APP/PS1 groups. Further findings indicate that enhanced microglial Aß phagocytosis and lower levels of proinflammatory cytokines were concurrent with the aforementioned effects of NP106 in combination with TML-6. Intriguingly, combination treatment also normalized the gut microbiota of APP/PS1 mice to levels resembling the wild-type control. Taken together, combination treatment outperformed NP106 or TML-6 monotherapy in ameliorating Aß pathology and the nesting behavioral deficit in APP/PS1 mice. The superior effect might result from a more potent modulation of microglial function, cerebral inflammation, and the gut microbiota. This innovative treatment paradigm confers a new avenue to develop more efficacious AD treatments.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/deficiência , Anticorpos Monoclonais/farmacologia , Curcumina/farmacologia , Presenilina-1/deficiência , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Biomarcadores , Curcumina/análogos & derivados , Gerenciamento Clínico , Modelos Animais de Doenças , Suscetibilidade a Doenças , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microbiota/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Terapia de Alvo Molecular , Placa Amiloide/tratamento farmacológico , Placa Amiloide/patologia
2.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34172567

RESUMO

Alterations in Ca2+ homeostasis have been reported in several in vitro and in vivo studies using mice expressing the Alzheimer's disease-associated transgenes, presenilin and the amyloid precursor protein (APP). While intense research focused on amyloid-ß-mediated functions on neuronal Ca2+ handling, the physiological role of APP and its close homolog APLP2 is still not fully clarified. We now elucidate a mechanism to show how APP and its homolog APLP2 control neuronal Ca2+ handling and identify especially the ectodomain APPsα as an essential regulator of Ca2+ homeostasis. Importantly, we demonstrate that the loss of APP and APLP2, but not APLP2 alone, impairs Ca2+ handling, the refill of the endoplasmic reticulum Ca2+ stores, and synaptic plasticity due to altered function and expression of the SERCA-ATPase and expression of store-operated Ca2+ channel-associated proteins Stim1 and Stim2. Long-term AAV-mediated expression of APPsα, but not acute application of the recombinant protein, restored physiological Ca2+ homeostasis and synaptic plasticity in APP/APLP2 cDKO cultures. Overall, our analysis reveals an essential role of the APP family and especially of the ectodomain APPsα in Ca2+ homeostasis, thereby highlighting its therapeutic potential.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Cálcio/metabolismo , Hipocampo/patologia , Homeostase , Neurônios/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Potenciais Pós-Sinápticos Excitadores , Integrases/metabolismo , Potenciação de Longa Duração , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Regulação para Cima
3.
Front Immunol ; 11: 1781, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849642

RESUMO

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of dementia in older adults. Although amyloid-beta (Aß) plaque deposition and chronic neuroinflammation in the central nervous system (CNS) contribute to AD pathology, neither Aß plaque removal nor anti-inflammatory therapy has shown much clinical success, suggesting that the combinational therapies for the disease-causative factors may be needed for amelioration. Recent data also suggest that systemic immunity in AD should be boosted, rather than suppressed, to drive an immune-dependent cascade needed for Aß clearance and brain repair. Thymic epithelial cells (TECs) not only play a critical role in supporting T cell development but also mediate the deletion of autoreactive T cells by expressing autoantigens. We have reported that embryonic stem cells (ESCs) can be selectively induced to differentiate into thymic epithelial progenitors (TEPs) in vitro that further develop into TECs in vivo to support T cell development. We show here that transplantation of mouse ESC (mESC)-TEPs into AD mice reduced cerebral Aß plaque load and improved cognitive performance, in correlation with an increased number of T cells, enhanced choroid plexus (CP) gateway activity, and increased number of macrophages in the brain. Furthermore, transplantation of the amyloid precursor protein (APP) gene deleted mESC-TEPs (APP-/-) results in more effective reduction of AD pathology as compared to wild-type (APP+/+) mESC-TEPs. This is associated with the generation of Aß-specific T cells, which leads to an increase of anti-Aß antibody (Ab)-producing B cells in the spleen and enhanced levels of anti-Aß antibodies in the serum, as well as an increase of Aß phagocytosing macrophages in the CNS. Our results suggest that transplantation of APP-/- human ESC- or induced pluripotent stem cell (iPSC)-derived TEPs may provide a new tool to mitigate AD in patients.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Células-Tronco Embrionárias/transplante , Células Epiteliais/transplante , Linfopoese/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Timo/imunologia
4.
Glia ; 67(5): 985-998, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30667091

RESUMO

The investigation of amyloid precursor protein (APP) has been mainly confined to its neuronal functions, whereas very little is known about its physiological role in astrocytes. Astrocytes exhibit a particular morphology with slender extensions protruding from somata and primary branches. Along these fine extensions, spontaneous calcium transients occur in spatially restricted microdomains. Within these microdomains mitochondria are responsible for local energy supply and Ca2+ buffering. Using two-photon in vivo Ca2+ imaging, we report a significant decrease in the density of active microdomains, frequency of spontaneous Ca2+ transients and slower Ca2+ kinetics in mice lacking APP. Mechanistically, these changes could be potentially linked to mitochondrial malfunction as our in vivo and in vitro data revealed severe, APP-dependent structural mitochondrial fragmentation in astrocytes. Functionally, such mitochondria exhibited prolonged kinetics and morphology dependent signal size of ATP-induced Ca2+ transients. Our results highlight a prominent role of APP in the modulation of Ca2+ activity in astrocytic microdomains whose precise functioning is crucial for the reinforcement and modulation of synaptic function. This study provides novel insights in APP physiological functions which are important for the understanding of the effects of drugs validated in Alzheimer's disease treatment that affect the function of APP.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Astrócitos/ultraestrutura , Encéfalo/citologia , Cálcio/metabolismo , Microdomínios da Membrana/metabolismo , Mitocôndrias/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/ultraestrutura , Transdução Genética , Transfecção
5.
Neurochem Res ; 44(6): 1346-1355, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29572646

RESUMO

The function of the ß-A4 amyloid protein precursor (APP) of Alzheimer's disease (AD) remains unclear. APP has a number of putative roles in neuronal differentiation, survival, synaptogenesis and cell adhesion. In this study, we examined the development of axons, dendrites and synapses in cultures of hippocampus neutrons derived from APP knockout (KO) mice. We report that loss of APP function reduces the branching of cultured hippocampal neurons, resulting in reduced synapse formation. Using a compartmentalised culture approach, we found reduced axonal outgrowth in cultured hippocampal neurons and we also identified abnormal growth characteristics of isolated hippocampal neuron axons. Although APP has previously been suggested to play an important role in promoting cell adhesion, we surprisingly found that APPKO hippocampal neurons adhered more strongly to a poly-L-lysine substrate and their neurites displayed an increased density of focal adhesion puncta. The findings suggest that the function of APP has an important role in both dendritic and axonal growth and that endogenous APP may regulate substrate adhesion of hippocampal neurons. The results may explain neuronal and synaptic morphological abnormalities in APPKO mice and the presence of abnormal APP expression in dystrophic neurites around amyloid deposits in AD.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Axônios/metabolismo , Dendritos/metabolismo , Hipocampo/metabolismo , Sinapses/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Adesão Celular/fisiologia , Feminino , Técnicas de Inativação de Genes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Crescimento Neuronal/fisiologia , Gravidez
6.
Neurochem Res ; 44(6): 1356-1366, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30362021

RESUMO

The amyloid precursor protein (APP) is a member of a conserved gene family that includes the amyloid precursor-like proteins 1 (APLP1) and 2 (APLP2). APP and APLP2 share a high degree of similarity, and have overlapping patterns of spatial and temporal expression in the central and peripheral tissues, in particular at the neuromuscular junction. APP-family knockout (KO) studies have helped elucidate aspects of function and functional redundancy amongst the APP-family members. In the present study, we investigated motor performance of APLP2-KO mice and the effect sex differences and age-related changes have on motor performance. APLP2-KO and WT (on C57Bl6 background) littermates control mice from 8 (young adulthood) to 48 weeks (middle age) were investigated. Analysis of motor neuron and muscle morphology showed APLP2-KO females but not males, had less age-related motor function impairments. We observed age and sex differences in both motor neuron number and muscle fiber size distribution for APLP2-KO mice compared to WT (C57Bl6). These alterations in the motor neuron number and muscle fiber distribution pattern may explain why female APLP2-KO mice have far better motor function behaviour during ageing.


Assuntos
Envelhecimento/fisiologia , Precursor de Proteína beta-Amiloide/deficiência , Atividade Motora/fisiologia , Fatores Etários , Envelhecimento/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/patologia , Músculo Esquelético/patologia , Fatores Sexuais , Medula Espinal/patologia
7.
Neurotherapeutics ; 15(4): 1055-1062, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30112699

RESUMO

Proteolytic cleavage of the amyloid precursor protein (APP) into the Aß peptide has been an extensively researched mechanism for Alzheimer's disease, but the normal function of the protein is less understood. APP functions to regulate neuronal iron content by stabilizing the surface presentation of ferroportin-the only iron exporter channel of cells. The present study aims to quantify the contribution of APP to brain and peripheral iron by examining the lifetime impact on brain and liver iron levels in APP knockout mice. Consistent with previous reports, we found that wild-type mice exhibited an age-dependent increase in iron and ferritin in the brain, while no age-dependent changes were observed in the liver. APP ablation resulted in an exaggeration of age-dependent iron accumulation in the brain and liver in mice that was assessed at 8, 12, 18, and 22 months of age. Brain ferroportin levels were decreased in APP knockout mice, consistent with a mechanistic role for APP in stabilizing this iron export protein in the brain. Iron elevation in the brain and liver of APP knockout mice correlated with decreased transferrin receptor 1 and increased ferritin protein levels. However, no age-dependent increase in brain ferritin iron saturation was observed in APP-KO mice despite similar protein expression levels potentially explaining the vulnerability of APP-KO mice to parkinsonism and traumatic brain sequelae. Our results support a crucial role of APP in regulating brain and peripheral iron, and show that APP may act to oppose brain iron elevation during aging.


Assuntos
Envelhecimento/patologia , Precursor de Proteína beta-Amiloide/deficiência , Encéfalo/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Fatores Etários , Precursor de Proteína beta-Amiloide/genética , Animais , Ferritinas/metabolismo , Camundongos , Camundongos Knockout
8.
Biol Chem ; 399(5): 453-465, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29337689

RESUMO

Employing hippocampal synaptosomes from amyloid precursor protein (APP)-deleted mice we analyzed the immediate effects of amyloid beta peptide 42 (Aß42) peptide in its oligomeric or fibrillar assembly or of soluble amyloid precursor protein alpha (sAPPα) protein on their bioenergetic activity. Upon administration of oligomeric Aß42 peptide for 30 min we observed a robust decrease both in mitochondrial activity and in mitochondrial membrane potential (MMP). In contrast the respective fibrillary or scrambled peptides showed no effect, indicating that inhibition strictly depends on the oligomerization status of the peptide. Hippocampal synaptosomes from old APP-KO mice revealed a further reduction of their already impaired bioenergetic activity upon incubation with 10 µm Aß42 peptide. In addition we evaluated the influence of the sAPPα protein on mitochondrial activity of hippocampal synaptosomes derived from young or old APP-KO animals. In neither case 20 nm nor 200 nm sAPPα protein had an effect on mitochondrial metabolic activity. Our findings demonstrate that hippocampal synaptosomes derived from APP-KO mice are a most suitable model system to evaluate the impact of Aß42 peptide on its bioenergetic activity and to further elucidate the molecular mechanisms underlying the impairments by oligomeric Aß42 on mitochondrial function. Our data demonstrate that extracellular Aß42 peptide is taken up into synaptosomes where it immediately attenuates mitochondrial activity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Sinaptossomos/metabolismo , Precursor de Proteína beta-Amiloide/deficiência , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo
9.
J Cereb Blood Flow Metab ; 38(10): 1715-1726, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-28959912

RESUMO

The exact physiological function of amyloid-ß precursor protein (APP) in endothelial cells is unknown. Endothelium-specific APP-deficient (eAPP-/-) mice were created to gain new insights into the role of APP in the control of vascular endothelial function. Endothelium-dependent relaxations to acetylcholine were significantly impaired in basilar arteries of global APP knockout (APP-/-) and eAPP-/- mice ( P < 0.05). In contrast, endothelium-independent relaxations to nitric oxide (NO)-donor diethylamine-NONOate were unchanged. Western blot analysis revealed that protein expression of endothelial nitric oxide synthase (eNOS) was significantly downregulated in large cerebral arteries of APP-/- mice and eAPP-/- mice as compared to respective wild-type littermates ( P < 0.05). Furthermore, basal levels of cyclic guanosine monophosphate (cGMP) were also significantly reduced in large cerebral arteries of APP-deficient mice ( P < 0.05). In contrast, protein expression of prostacyclin synthase as well as levels of cyclic adenosine monophosphate (cAMP) was not affected by genetic inactivation of APP in endothelial cells. By using siRNA to knockdown APP in cultured human brain microvascular endothelial cells we also found a significant downregulation of eNOS mRNA and protein expressions in APP-deficient endothelium ( P < 0.05). These findings indicate that under physiological conditions, expression of APP in cerebral vascular endothelium plays an important protective function by maintaining constitutive expression of eNOS .


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Artérias Cerebrais/metabolismo , Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Células Cultivadas , Endotélio Vascular/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
Brain Struct Funct ; 223(1): 267-284, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28779307

RESUMO

The locus coeruleus (LC)-norepinephrine (NE) system is an understudied circuit in the context of Alzheimer's disease (AD), and is thought to play an important role in neurodegenerative and neuropsychiatric diseases involving catecholamine neurotransmitters. Understanding the expression and distribution of the amyloid beta (Aß) peptide, a primary component of AD, under basal conditions and under conditions of NE perturbation within the coeruleo-cortical pathway may be important for understanding its putative role in pathological states. Thus, the goal of this study is to define expression levels and the subcellular distribution of endogenous Aß with respect to noradrenergic profiles in the rodent LC and medial prefrontal cortex (mPFC) and, further, to determine the functional relevance of NE in modulating endogenous Aß42 levels. We report that endogenous Aß42 is localized to tyrosine hydroxylase (TH) immunoreactive somatodendritic profiles of the LC and dopamine-ß-hydroxylase (DßH) immunoreactive axon terminals of the infralimbic mPFC (ILmPFC). Male and female naïve rats have similar levels of amyloid precursor protein (APP) cleavage products demonstrated by western blot, as well as similar levels of endogenous Aß42 as determined by enzyme-linked immunosorbent assay. Two models of NE depletion, DSP-4 lesion and DßH knockout (KO) mice, were used to assess the functional relevance of NE on endogenous Aß42 levels. DSP-4 lesioned rats and DßH-KO mice show significantly lower levels of endogenous Aß42. Noradrenergic depletion did not change APP-cleavage products resulting from ß-secretase processing. Thus, resultant decreases in endogenous Aß42 may be due to decreased neuronal activity of noradrenergic neurons, or, by decreased stimulation of adrenergic receptors which are known to contribute to Aß42 production by enhancing γ-secretase processing under normal physiological conditions.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/metabolismo , Locus Cerúleo/metabolismo , Norepinefrina/deficiência , Fragmentos de Peptídeos/metabolismo , Proteína ADAM10/metabolismo , Neurônios Adrenérgicos/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/metabolismo , Benzilaminas/toxicidade , Córtex Cerebral/ultraestrutura , Dopamina beta-Hidroxilase/deficiência , Dopamina beta-Hidroxilase/genética , Dopamina beta-Hidroxilase/ultraestrutura , Feminino , Locus Cerúleo/ultraestrutura , Masculino , Camundongos Knockout , Microscopia Eletrônica , Vias Neurais/metabolismo , Inibidores da Captação de Neurotransmissores/toxicidade , Fragmentos de Peptídeos/ultraestrutura , Ratos , Ratos Sprague-Dawley , Frações Subcelulares/metabolismo , Frações Subcelulares/ultraestrutura , Tirosina 3-Mono-Oxigenase/metabolismo
11.
J Neurosci ; 37(49): 11947-11966, 2017 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-29101243

RESUMO

Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD) and several theories have been advanced to explain the relationship. A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid ß-protein (Aß), self-associates to form soluble aggregates that impair synaptic and network activity. Here, we used the most disease-relevant form of Aß, protein isolated from AD brain. Using this material, we show that the synaptotoxic effects of Aß depend on expression of APP and that the Aß-mediated impairment of synaptic plasticity is accompanied by presynaptic effects that disrupt the excitatory/inhibitory (E/I) balance. The net increase in the E/I ratio and inhibition of plasticity are associated with Aß localizing to synapses and binding of soluble Aß aggregates to synapses requires the expression of APP. Our findings indicate a role for APP in AD pathogenesis beyond the generation of Aß and suggest modulation of APP expression as a therapy for AD.SIGNIFICANCE STATEMENT Here, we report on the plasticity-disrupting effects of amyloid ß-protein (Aß) isolated from Alzheimer's disease (AD) brain and the requirement of amyloid precursor protein (APP) for these effects. We show that Aß-containing AD brain extracts block hippocampal LTP, augment glutamate release probability, and disrupt the excitatory/inhibitory balance. These effects are associated with Aß localizing to synapses and genetic ablation of APP prevents both Aß binding and Aß-mediated synaptic dysfunctions. Our results emphasize the importance of APP in AD and should stimulate new studies to elucidate APP-related targets suitable for pharmacological manipulation.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/biossíntese , Encéfalo/metabolismo , Plasticidade Neuronal/fisiologia , Fragmentos de Peptídeos/metabolismo , Sinapses/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Animais , Encéfalo/patologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Ligação Proteica/fisiologia , Sinapses/patologia
12.
Sci Rep ; 7(1): 14512, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29109429

RESUMO

Familial Alzheimer's disease (AD) is caused by mutations in the genes that encode amyloid precursor protein (APP) and presenilins. Disturbances in calcium homeostasis have been observed in various cellular and animal models of AD and are proposed to underlie the pathogenesis of the disease. Furthermore, wildtype presenilins were shown to regulate endoplasmic reticulum (ER) calcium homeostasis, although their precise mechanism of action remains controversial. To investigate whether APP also affects ER calcium levels, we used RNA interference to target the APP gene in cultured T84 cells in combination with two types of ER calcium sensors. Using a genetically encoded calcium indicator, GEM-CEPIA1er, we found that APP-deficient cells exhibited elevated resting calcium levels in the ER and prolonged emptying of ER calcium stores upon the cyclopiazonic acid-induced inhibition of sarco-endoplasmic reticulum calcium-ATPase. These effects could be ascribed to lower ER calcium leakage rates. Consistent with these results, translocation of the endogenous ER calcium sensor STIM1 to its target channel Orai1 was delayed following ER calcium store depletion. Our data suggest a physiological function of APP in the regulation of ER calcium levels.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Precursor de Proteína beta-Amiloide/genética , Transporte Biológico , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HEK293 , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Indóis/farmacologia , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Molécula 1 de Interação Estromal/metabolismo
13.
Glia ; 65(4): 569-580, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28130845

RESUMO

Astrocytes play crucial roles in brain homeostasis and are emerging as regulatory elements of neuronal and synaptic physiology by responding to neurotransmitters with Ca2+ elevations and releasing gliotransmitters that activate neuronal receptors. Aging involves neuronal and astrocytic alterations, being considered risk factor for neurodegenerative diseases. Most evidence of the astrocyte-neuron signaling is derived from studies with young animals; however, the features of astrocyte-neuron signaling in adult and aging brain remain largely unknown. We have investigated the existence and properties of astrocyte-neuron signaling in physiologically and pathologically aging mouse hippocampal and cortical slices at different lifetime points (0.5 to 20 month-old animals). We found that astrocytes preserved their ability to express spontaneous and neurotransmitter-dependent intracellular Ca2+ signals from juvenile to aging brains. Likewise, resting levels of gliotransmission, assessed by neuronal NMDAR activation by glutamate released from astrocytes, were largely preserved with similar properties in all tested age groups, but DHPG-induced gliotransmission was reduced in aged mice. In contrast, gliotransmission was enhanced in the APP/PS1 mouse model of Alzheimer's disease, indicating a dysregulation of astrocyte-neuron signaling in pathological conditions. Disruption of the astrocytic IP3 R2 mediated-signaling, which is required for neurotransmitter-induced astrocyte Ca2+ signals and gliotransmission, boosted the progression of amyloid plaque deposits and synaptic plasticity impairments in APP/PS1 mice at early stages of the disease. Therefore, astrocyte-neuron interaction is a fundamental signaling, largely conserved in the adult and aging brain of healthy animals, but it is altered in Alzheimer's disease, suggesting that dysfunctions of astrocyte Ca2+ physiology may contribute to this neurodegenerative disease. GLIA 2017 GLIA 2017;65:569-580.


Assuntos
Envelhecimento , Astrócitos/fisiologia , Encéfalo/citologia , Comunicação Celular/fisiologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Acetilcolina/farmacologia , Trifosfato de Adenosina/farmacologia , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Astrócitos/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Cálcio/metabolismo , Comunicação Celular/efeitos dos fármacos , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Presenilina-1/deficiência , Presenilina-1/genética , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/genética
14.
Nature ; 552(7685): 355-361, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29293211

RESUMO

The spreading of pathology within and between brain areas is a hallmark of neurodegenerative disorders. In patients with Alzheimer's disease, deposition of amyloid-ß is accompanied by activation of the innate immune system and involves inflammasome-dependent formation of ASC specks in microglia. ASC specks released by microglia bind rapidly to amyloid-ß and increase the formation of amyloid-ß oligomers and aggregates, acting as an inflammation-driven cross-seed for amyloid-ß pathology. Here we show that intrahippocampal injection of ASC specks resulted in spreading of amyloid-ß pathology in transgenic double-mutant APPSwePSEN1dE9 mice. By contrast, homogenates from brains of APPSwePSEN1dE9 mice failed to induce seeding and spreading of amyloid-ß pathology in ASC-deficient APPSwePSEN1dE9 mice. Moreover, co-application of an anti-ASC antibody blocked the increase in amyloid-ß pathology in APPSwePSEN1dE9 mice. These findings support the concept that inflammasome activation is connected to seeding and spreading of amyloid-ß pathology in patients with Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Microglia/metabolismo , Agregação Patológica de Proteínas , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Anticorpos/farmacologia , Proteínas Adaptadoras de Sinalização CARD/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização CARD/química , Proteínas Adaptadoras de Sinalização CARD/imunologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Presenilina-1/deficiência , Presenilina-1/genética , Domínios Proteicos , Memória Espacial/fisiologia
15.
Neuropharmacology ; 110(Pt A): 493-502, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27553120

RESUMO

Though amyloid precursor protein (APP) can potentially be cleaved to generate the pathological amyloid ß peptide (Aß), APP itself plays an important role in regulating neuronal activity. APP deficiency causes functional impairment in cholinergic synaptic transmission and cognitive performance. However, the mechanisms underlying altered cholinergic synaptic transmission in APP knock-out mice (APP(-/-)) are poorly understood. In this study, we conducted in vivo extracellular recording to investigate cholinergic compound action potentials (CAPs) of the superior cervical ganglion (SCG) in APP(-/-) and littermate wild-type (WT) mice. Our results demonstrate that APP not only regulates presynaptic activity, but also affects postsynaptic function at cholinergic synapses in SCG. APP deficiency reduces the number of vesicles in presynaptic terminalsand attenuatesthe amplitude of CAPs, likely due to dysfunction of high-affinity choline transporters. Pharmacological and biochemical examination showed that postsynaptic responsesmediated by α4ß2 and α7 nicotinic acetylcholine receptors are reduced in the absence of APP. Our research provides evidences on how APP regulates cholinergic function and therefore may help to identify potential therapeutic targets to treat cholinergic dysfunction associated with Alzheimer's disease pathogenesis.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Receptores Nicotínicos/metabolismo , Gânglio Cervical Superior/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Acetilcolina/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Colinérgicos/farmacologia , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Camundongos Knockout , Gânglio Cervical Superior/efeitos dos fármacos , Gânglio Cervical Superior/patologia , Sinapses/patologia , Transmissão Sináptica/efeitos dos fármacos
16.
Mol Brain ; 9(1): 64, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27267879

RESUMO

BACKGROUND: Amyloid precursor protein knockout mice (APP-KO) have impaired differentiation of amacrine and horizontal cells. APP is part of a gene family and its paralogue amyloid precursor-like protein 2 (APLP2) has both shared as well as distinct expression patterns to APP, including in the retina. Given the impact of APP in the retina we investigated how APLP2 expression affected the retina using APLP2 knockout mice (APLP2-KO). RESULTS: Using histology, morphometric analysis with noninvasive imaging technique and electron microscopy, we showed that APLP2-KO retina displayed abnormal formation of the outer synaptic layer, accompanied with greatly impaired photoreceptor ribbon synapses in adults. Moreover, APLP2-KO displayed a significant decease in ON-bipolar, rod bipolar and type 2 OFF-cone bipolar cells (36, 21 and 63 %, respectively). Reduction of the number of bipolar cells was accompanied with disrupted dendrites, reduced expression of metabotropic glutamate receptor 6 at the dendritic tips and alteration of axon terminals in the OFF laminae of the inner plexiform layer. In contrast, the APP-KO photoreceptor ribbon synapses and bipolar cells were intact. The APLP2-KO retina displayed numerous phenotypic similarities with the congenital stationary night blindness, a non-progressive retinal degeneration disease characterized by the loss of night vision. The pathological phenotypes in the APLP2-KO mouse correlated to altered transcription of genes involved in pre- and postsynatic structure/function, including CACNA1F, GRM6, TRMP1 and Gα0, and a normal scotopic a-wave electroretinogram amplitude, markedly reduced scotopic electroretinogram b-wave and modestly reduced photopic cone response. This confirmed the impaired function of the photoreceptor ribbon synapses and retinal bipolar cells, as is also observed in congenital stationary night blindness. Since congenital stationary night blindness present at birth, we extended our analysis to retinal differentiation and showed impaired differentiation of different bipolar cell subtypes and an altered temporal sequence of development from OFF to ON laminae in the inner plexiform layer. This was associated with the altered expression patterns of bipolar cell generation and differentiation factors, including MATH3, CHX10, VSX1 and OTX2. CONCLUSIONS: These findings demonstrate that APLP2 couples retina development and synaptic genes and present the first evidence that APLP2 expression may be linked to synaptic disease.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Oftalmopatias Hereditárias/genética , Deleção de Genes , Doenças Genéticas Ligadas ao Cromossomo X/genética , Miopia/genética , Cegueira Noturna/genética , Envelhecimento/patologia , Células Amácrinas/metabolismo , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proteínas do Sistema Complemento/metabolismo , Dendritos/metabolismo , Oftalmopatias Hereditárias/patologia , Oftalmopatias Hereditárias/fisiopatologia , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miopia/patologia , Miopia/fisiopatologia , Neurogênese , Cegueira Noturna/patologia , Cegueira Noturna/fisiopatologia , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Células Fotorreceptoras de Vertebrados/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Bipolares da Retina/metabolismo , Células Bipolares da Retina/patologia , Células Bipolares da Retina/ultraestrutura , Transmissão Sináptica , Fatores de Transcrição/metabolismo , Transcrição Gênica
17.
PLoS Comput Biol ; 12(4): e1004832, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27092780

RESUMO

The hallmarks of Alzheimer's disease (AD) are characterized by cognitive decline and behavioral changes. The most prominent brain region affected by the progression of AD is the hippocampal formation. The pathogenesis involves a successive loss of hippocampal neurons accompanied by a decline in learning and memory consolidation mainly attributed to an accumulation of senile plaques. The amyloid precursor protein (APP) has been identified as precursor of Aß-peptides, the main constituents of senile plaques. Until now, little is known about the physiological function of APP within the central nervous system. The allocation of APP to the proteome of the highly dynamic presynaptic active zone (PAZ) highlights APP as a yet unknown player in neuronal communication and signaling. In this study, we analyze the impact of APP deletion on the hippocampal PAZ proteome. The native hippocampal PAZ derived from APP mouse mutants (APP-KOs and NexCreAPP/APLP2-cDKOs) was isolated by subcellular fractionation and immunopurification. Subsequently, an isobaric labeling was performed using TMT6 for protein identification and quantification by high-resolution mass spectrometry. We combine bioinformatics tools and biochemical approaches to address the proteomics dataset and to understand the role of individual proteins. The impact of APP deletion on the hippocampal PAZ proteome was visualized by creating protein-protein interaction (PPI) networks that incorporated APP into the synaptic vesicle cycle, cytoskeletal organization, and calcium-homeostasis. The combination of subcellular fractionation, immunopurification, proteomic analysis, and bioinformatics allowed us to identify APP as structural and functional regulator in a context-sensitive manner within the hippocampal active zone network.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Biologia Computacional , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terminações Pré-Sinápticas/metabolismo , Mapas de Interação de Proteínas , Proteoma/metabolismo , Sinapses/metabolismo
18.
Sci Rep ; 6: 21948, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26905287

RESUMO

Amyloid precursor protein (APP) is critically involved in the pathophysiology of Alzheimer's disease, but its physiological functions remain elusive. Importantly, APP knockout (APP-KO) mice exhibit cognitive deficits, suggesting that APP plays a role at the neuronal network level. To investigate this possibility, we recorded local field potentials (LFPs) from the posterior parietal cortex, dorsal hippocampus and lateral prefrontal cortex of freely moving APP-KO mice. Spectral analyses showed that network oscillations within the theta- and gamma-frequency bands were not different between APP-KO and wild-type mice. Surprisingly, however, while gamma amplitude coupled to theta phase in all recorded regions of wild-type animals, in APP-KO mice theta-gamma coupling was strongly diminished in recordings from the parietal cortex and hippocampus, but not in LFPs recorded from the prefrontal cortex. Thus, lack of APP reduces oscillatory coupling in LFP recordings from specific brain regions, despite not affecting the amplitude of the oscillations. Together, our findings reveal reduced cross-frequency coupling as a functional marker of APP deficiency at the network level.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Disfunção Cognitiva/fisiopatologia , Ritmo Gama , Hipocampo/fisiopatologia , Lobo Parietal/fisiopatologia , Ritmo Teta , Precursor de Proteína beta-Amiloide/genética , Animais , Disfunção Cognitiva/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rede Nervosa/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Técnicas Estereotáxicas
19.
Nat Commun ; 6: 8997, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26646366

RESUMO

Alzheimer's disease (AD) is characterized by the transition of amyloid-ß (Aß) monomers into toxic oligomers and plaques. Given that Aß abnormality typically precedes the development of clinical symptoms, an agent capable of disaggregating existing Aß aggregates may be advantageous. Here we report that a small molecule, 4-(2-hydroxyethyl)-1-piperazinepropanesulphonic acid (EPPS), binds to Aß aggregates and converts them into monomers. The oral administration of EPPS substantially reduces hippocampus-dependent behavioural deficits, brain Aß oligomer and plaque deposits, glial γ-aminobutyric acid (GABA) release and brain inflammation in an Aß-overexpressing, APP/PS1 transgenic mouse model when initiated after the development of severe AD-like phenotypes. The ability of EPPS to rescue Aß aggregation and behavioural deficits provides strong support for the view that the accumulation of Aß is an important mechanism underlying AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/deficiência , Hipocampo/efeitos dos fármacos , Piperazinas/administração & dosagem , Placa Amiloide/metabolismo , Presenilina-1/deficiência , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/genética , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Presenilina-1/genética
20.
PLoS One ; 10(11): e0143135, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26606074

RESUMO

OBJECTIVE: This study investigated the effects of intervention with a combination of nutrients in the amyloid precursor protein-presenilin (APP-PSN) C57BL/6J double transgenic mouse model of Alzheimer's disease (AD). METHODS: A total of 72 2-month-old APP-PSN mice were randomly assigned to three groups. The model group (MG) was fed regular, unsupplemented chow, while the low- and high-dose treatment groups (LG and HG, respectively) were given a combination of nutrients that included phosphatidylserine, blueberry extracts, docosahexaenoic acid, and eicosapentaenoic acid as part of their diet. An additional 24 wild-type littermates that were fed unsupplemented chow served as the negative control group (NG). After 3 and 7 months of treatment, the cognitive performance was assessed with the Morris water maze and the shuttle box escape/avoidance task, and the biochemical parameters and oxidative stress were evaluated in both the blood and brain. RESULTS: An improvement in antioxidant capacity was observed in the treatment groups relative to the MG at 3 months, while superior behavioral test results were observed in the mice of the HG and NG groups. In the MG, pycnosis was detected in neuronal nuclei, and a loss of neurons was observed in the cerebral cortex and the hippocampus. At 7 months, the ß-amyloid1-42 peptide accumulation was significantly elevated in the MG but was markedly lower in the mice fed the nutrient combination. The antioxidant capacity and behavioral test scores were also higher in these mice. CONCLUSIONS: Early intervention with a combination of nutrients should be considered as a strategy for preventing cognitive decline and other symptoms associated with AD.


Assuntos
Doença de Alzheimer/genética , Ração Animal , Suplementos Nutricionais , Acetilcolina/sangue , Acetilcolina/metabolismo , Doença de Alzheimer/dietoterapia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal , Peso Corporal , Colinesterases/sangue , Colinesterases/metabolismo , Modelos Animais de Doenças , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estresse Oxidativo , Presenilinas/deficiência , Presenilinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...