Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Traffic ; 25(5): e12937, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38777335

RESUMO

The polymorphic APOE gene is the greatest genetic determinant of sporadic Alzheimer's disease risk: the APOE4 allele increases risk, while the APOE2 allele is neuroprotective compared with the risk-neutral APOE3 allele. The neuronal endosomal system is inherently vulnerable during aging, and APOE4 exacerbates this vulnerability by driving an enlargement of early endosomes and reducing exosome release in the brain of humans and mice. We hypothesized that the protective effects of APOE2 are, in part, mediated through the endosomal pathway. Messenger RNA analyses showed that APOE2 leads to an enrichment of endosomal pathways in the brain when compared with both APOE3 and APOE4. Moreover, we show age-dependent alterations in the recruitment of key endosomal regulatory proteins to vesicle compartments when comparing APOE2 to APOE3. In contrast to the early endosome enlargement previously shown in Alzheimer's disease and APOE4 models, we detected similar morphology and abundance of early endosomes and retromer-associated vesicles within cortical neurons of aged APOE2 targeted-replacement mice compared with APOE3. Additionally, we observed increased brain extracellular levels of endosome-derived exosomes in APOE2 compared with APOE3 mice during aging, consistent with enhanced endosomal cargo clearance by exosomes to the extracellular space. Our findings thus demonstrate that APOE2 enhances an endosomal clearance pathway, which has been shown to be impaired by APOE4 and which may be protective due to APOE2 expression during brain aging.


Assuntos
Envelhecimento , Apolipoproteína E2 , Encéfalo , Endossomos , Exossomos , Animais , Humanos , Camundongos , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E2/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E4/genética , Encéfalo/metabolismo , Endossomos/metabolismo , Exossomos/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo
2.
Cell Mol Life Sci ; 81(1): 224, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769196

RESUMO

Synaptic loss is an early event in the penumbra area after an ischemic stroke. Promoting synaptic preservation in this area would likely improve functional neurological recovery. We aimed to detect proteins involved in endogenous protection mechanisms of synapses in the penumbra after stroke and to analyse potential beneficial effects of these candidates for a prospective stroke treatment. For this, we performed Liquid Chromatography coupled to Mass Spectrometry (LC-MS)-based proteomics of synaptosomes isolated from the ipsilateral hemispheres of mice subjected to experimental stroke at different time points (24 h, 4 and 7 days) and compared them to sham-operated mice. Proteomic analyses indicated that, among the differentially expressed proteins between the two groups, cystatin C (CysC) was significantly increased at 24 h and 4 days following stroke, before returning to steady-state levels at 7 days, thus indicating a potential transient and intrinsic rescue mechanism attempt of neurons. When CysC was applied to primary neuronal cultures subjected to an in vitro model of ischemic damage, this treatment significantly improved the preservation of synaptic structures. Notably, similar effects were observed when CysC was loaded into brain-derived extracellular vesicles (BDEVs). Finally, when CysC contained in BDEVs was administered intracerebroventricularly to stroked mice, it significantly increased the expression of synaptic markers such as SNAP25, Homer-1, and NCAM in the penumbra area compared to the group supplied with empty BDEVs. Thus, we show that CysC-loaded BDEVs promote synaptic protection after ischemic damage in vitro and in vivo, opening the possibility of a therapeutic use in stroke patients.


Assuntos
Isquemia Encefálica , Encéfalo , Cistatina C , Vesículas Extracelulares , Camundongos Endogâmicos C57BL , Sinapses , Animais , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/transplante , Cistatina C/metabolismo , Sinapses/metabolismo , Camundongos , Masculino , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Proteômica/métodos , Sinaptossomos/metabolismo , Neurônios/metabolismo , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/terapia , Células Cultivadas , Modelos Animais de Doenças
3.
Neurochem Res ; 47(11): 3428-3439, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35904699

RESUMO

Extracellular vesicles (EVs) in the brain play a role in neuronal homeostasis by removing intracellular material and regulating cell-to-cell communication. Given that sex and aging differentially modulate brain networks, we investigated sex-dependent differences in EV levels and content in the brain during aging. EVs were isolated from the brains of 3, 6, 12, 18, and 24 month-old female and male C57BL/6 J mice, and the levels of different EV species determined. While the number of plasma membrane-derived microvesicles and a subset of late endosomes-derived exosomes increased with age in the brain of female mice, no significant changes were seen in males. Mitochondria-derived mitovesicles in the brain increased during aging in both sexes, a change that may reflect aging-dependent alterations in mitochondrial function. These findings reveal enhanced turnover during aging in female brains, suggesting a mechanism for advantageous successful female brain aging and sex-depending different susceptibility to age-related neurodegenerative diseases.


Assuntos
Exossomos , Vesículas Extracelulares , Animais , Encéfalo , Feminino , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
Neurochem Res ; 47(8): 2263-2277, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35501523

RESUMO

In multiple neurodevelopmental and neurodegenerative disorders, endosomal changes correlate with changes in exosomes. We examined this linkage in the brain of mice that received cocaine injections for two weeks starting at 2.5 months of age. Cocaine caused a decrease in the number of both neuronal early and late endosomes and exosomes in the brains of male but not female mice. The response to cocaine in ovariectomized females mirrored male, demonstrating that these sex-differences in response to cocaine are driven by hormonal differences. Moreover, cocaine increased the amount of α-synuclein per exosome in the brain of females but did not affect exosomal α-synuclein content in the brain of males, a sex-difference eliminated by ovariectomy. Enhanced packaging of α-synuclein into female brain exosomes with the potential for propagation of pathology throughout the brain suggests a mechanism for the different response of females to chronic cocaine exposure as compared to males.


Assuntos
Cocaína , Exossomos , Animais , Cocaína/farmacologia , Endossomos , Feminino , Masculino , Camundongos , Neurônios/patologia , alfa-Sinucleína
5.
Sensors (Basel) ; 22(11)2022 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-35684879

RESUMO

Radar systems are mainly used for tracking aircraft, missiles, satellites, and watercraft. In many cases, information regarding the objects detected by a radar system is sent to, and used by, a peripheral consuming system, such as a missile system or a graphical user interface used by an operator. Those systems process the data stream and make real-time operational decisions based on the data received. Given this, the reliability and availability of information provided by radar systems have grown in importance. Although the field of cyber security has been continuously evolving, no prior research has focused on anomaly detection in radar systems. In this paper, we present an unsupervised deep-learning-based method for detecting anomalies in radar system data streams; we take into consideration the fact that a data stream created by a radar system is heterogeneous, i.e., it contains both numerical and categorical features with non-linear and complex relationships. We propose a novel technique that learns the correlation between numerical features and an embedding representation of categorical features in an unsupervised manner. The proposed technique, which allows for the detection of the malicious manipulation of critical fields in a data stream, is complemented by a timing-interval anomaly-detection mechanism proposed for the detection of message-dropping attempts. Real radar system data were used to evaluate the proposed method. Our experiments demonstrated the method's high detection accuracy on a variety of data-stream manipulation attacks (an average detection rate of 88% with a false -alarm rate of 1.59%) and message-dropping attacks (an average detection rate of 92% with a false-alarm rate of 2.2%).

6.
FASEB J ; 34(9): 12922-12931, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32772431

RESUMO

Pleiotropic roles are proposed for brain extracellular vesicles (EVs) in the development of Alzheimer's disease (AD). Our previous studies have suggested a beneficial role for EVs in AD, where the endosomal system in vulnerable neurons is compromised, contributing to the removal of accumulated material from neurons. However, the involvement of EVs in propagating AD amyloidosis throughout the brain has been considered because the amyloid-ß precursor protein (APP), APP metabolites, and key APP cleaving enzymes were identified in association with EVs. Here, we undertook to determine whether the secretase machinery is actively processing APP in EVs isolated from the brains of wild-type and APP overexpressing Tg2576 mice. We found that full-length APP is cleaved in EVs incubated in the absence of cells. The resulting metabolites, both α- and ß-APP carboxyl-terminal fragments and APP intracellular domain accumulate in EVs over time and amyloid-ß dimerizes. Thus, EVs contribute to the removal from neurons and transport of APP-derived neurotoxic peptides. While this is potentially a venue for propagation of the pathology throughout the brain, it may contribute to efficient removal of neurotoxic peptides from the brain.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo , Vesículas Extracelulares/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Agregação Patológica de Proteínas
7.
Neurochem Res ; 46(11): 2909-2922, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34245421

RESUMO

Cocaine is a highly addictive stimulant with diverse effects on physiology. Recent studies indicate the involvement of extracellular vesicles (EVs) secreted by neural cells in the cocaine addiction process. It is hypothesized that cocaine affects secretion levels of EVs and their cargos, resulting in modulation of synaptic transmission and plasticity related to addiction physiology and pathology. Lipids present in EVs are important for EV formation and for intercellular lipid exchange that may trigger physiological and pathological responses, including neuroplasticity, neurotoxicity, and neuroinflammation. Specific lipids are highly enriched in EVs compared to parent cells, and recent studies suggest the involvement of various lipids in drug-induced synaptic plasticity during the development and maintenance of addiction processes. Therefore, we examined interstitial small EVs isolated from the brain of mice treated with either saline or cocaine, focusing on the effects of cocaine on the lipid composition of EVs. We demonstrate that 12 days of noncontingent repeated cocaine (10 mg/kg) injections to mice, which induce locomotor sensitization, cause lipid composition changes in brain EVs of male mice as compared with saline-injected controls. The most prominent change is the elevation of GD1a ganglioside in brain EVs of males. However, cocaine does not affect the EV lipid profiles of the brain in female mice. Understanding the relationship between lipid composition in EVs and vulnerability to cocaine addiction may provide insight into novel targets for therapies for addiction.


Assuntos
Encéfalo/efeitos dos fármacos , Cocaína/toxicidade , Inibidores da Captação de Dopamina/toxicidade , Vesículas Extracelulares/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Caracteres Sexuais , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Feminino , Injeções Intraperitoneais , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
FASEB J ; 33(3): 3758-3771, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30481490

RESUMO

Accumulating evidence suggests that the abnormal aggregation of amyloid-ß (Αß) peptide in Alzheimer's disease (AD) begins intraneuronally, within vesicles of the endosomal-lysosomal pathway where Aß is both generated and degraded. Metalloproteases, including endothelin-converting enzyme (ECE)-1 and -2, reside within these vesicles and normally limit the accumulation of intraneuronally produced Aß. In this study, we determined whether disruption of Aß catabolism could trigger Aß aggregation within neurons and increase the amount of Aß associated with exosomes, small extracellular vesicles derived from endosomal multivesicular bodies. Using cultured cell lines, primary neurons, and organotypic brain slices from an AD mouse model, we found that pharmacological inhibition of the ECE family of metalloproteases increased intracellular and extracellular Aß levels and promoted the intracellular formation of Aß oligomers, a process that did not require internalization of secreted Aß. In vivo, the accumulation of intraneuronal Aß aggregates was accompanied by increased levels of both extracellular and exosome-associated Aß, including oligomeric species. Neuronal exosomes were found to contain both ECE-1 and -2 activities, suggesting that multivesicular bodies are intracellular sites of Aß degradation by these enzymes. ECE dysfunction could lead to the accumulation of intraneuronal Aß aggregates and their subsequent release into the extracellular space via exosomes.-Pacheco-Quinto, J., Clausen, D., Pérez-González, R., Peng, H., Meszaros, A., Eckman, C. B., Levy, E., Eckman, E. A. Intracellular metalloprotease activity controls intraneuronal Aß aggregation and limits secretion of Aß via exosomes.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Exossomos/metabolismo , Metaloendopeptidases/metabolismo , Agregação Patológica de Proteínas/metabolismo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Endossomos/metabolismo , Enzimas Conversoras de Endotelina/metabolismo , Espaço Extracelular/metabolismo , Feminino , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Corpos Multivesiculares/metabolismo , Neurônios/metabolismo , Proteólise
10.
Brain ; 142(1): 163-175, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30496349

RESUMO

In addition to being the greatest genetic risk factor for Alzheimer's disease, expression of the ɛ4 allele of apolipoprotein E can lead to cognitive decline during ageing that is independent of Alzheimer's amyloid-ß and tau pathology. In human post-mortem tissue and mouse models humanized for apolipoprotein E, we examined the impact of apolipoprotein E4 expression on brain exosomes, vesicles that are produced within and secreted from late-endocytic multivesicular bodies. Compared to humans or mice homozygous for the risk-neutral ɛ3 allele we show that the ɛ4 allele, whether homozygous or heterozygous with an ɛ3 allele, drives lower exosome levels in the brain extracellular space. In mice, we show that the apolipoprotein E4-driven change in brain exosome levels is age-dependent: while not present at age 6 months, it is detectable at 12 months of age. Expression levels of the exosome pathway regulators tumor susceptibility gene 101 (TSG101) and Ras-related protein Rab35 (RAB35) were found to be reduced in the brain at the protein and mRNA levels, arguing that apolipoprotein E4 genotype leads to a downregulation of exosome biosynthesis and release. Compromised exosome production is likely to have adverse effects, including diminishing a cell's ability to eliminate materials from the endosomal-lysosomal system. This reduction in brain exosome levels in 12-month-old apolipoprotein E4 mice occurs earlier than our previously reported brain endosomal pathway changes, arguing that an apolipoprotein E4-driven failure in exosome production plays a primary role in endosomal and lysosomal deficits that occur in apolipoprotein E4 mouse and human brains. Disruption of these interdependent endosomal-exosomal-lysosomal systems in apolipoprotein E4-expressing individuals may contribute to amyloidogenic amyloid-ß precursor protein processing, compromise trophic signalling and synaptic function, and interfere with a neuron's ability to degrade material, all of which are events that lead to neuronal vulnerability and higher risk of Alzheimer's disease development. Together, these data suggest that exosome pathway dysfunction is a previously unappreciated component of the brain pathologies that occur as a result of apolipoprotein E4 expression.


Assuntos
Apolipoproteína E4/biossíntese , Encéfalo/metabolismo , Exossomos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Alelos , Animais , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Proteínas de Ligação a DNA/biossíntese , Regulação para Baixo , Complexos Endossomais de Distribuição Requeridos para Transporte/biossíntese , Exossomos/ultraestrutura , Espaço Extracelular/metabolismo , Feminino , Genótipo , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fatores de Transcrição/biossíntese , Proteínas rab de Ligação ao GTP/biossíntese
11.
Neurobiol Dis ; 120: 165-173, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30176349

RESUMO

Cystatin C (CysC) plays diverse protective roles under conditions of neuronal challenge. We investigated whether CysC protects from trisomy-induced pathologies in a mouse model of Down syndrome (DS), the most common cause of developmental cognitive and behavioral impairments in humans. We have previously shown that the segmental trisomy mouse model, Ts[Rb(12.1716)]2Cje (Ts2) has DS-like neuronal and behavioral deficiencies. The current study reveals that transgene-mediated low levels of human CysC overexpression has a preventive effect on numerous neuropathologies in the brains of Ts2 mice, including reducing early and late endosome enlargement in cortical neurons and decreasing loss of basal forebrain cholinergic neurons (BFCNs). Consistent with these cellular benefits, behavioral dysfunctions were also prevented, including deficits in nesting behavior and spatial memory. We determined that the CysC-induced neuroprotective mechanism involves activation of the phosphotidylinositol kinase (PI3K)/AKT pathway. Activating this pathway leads to enhanced clearance of accumulated endosomal substrates, protecting cells from DS-mediated dysfunctions in the endosomal system and, for BFCNs, from neurodegeneration. Our findings suggest that modulation of the PI3/AKT pathway offers novel therapeutic interventions for patients with DS.


Assuntos
Cistatina C/biossíntese , Modelos Animais de Doenças , Síndrome de Down/metabolismo , Endossomos/metabolismo , Transdução de Sinais/fisiologia , Animais , Cistatina C/genética , Síndrome de Down/genética , Endossomos/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo
13.
PLoS Pathog ; 11(7): e1005032, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26154133

RESUMO

Recent studies have found that extracellular vesicles (EVs) play an important role in normal and disease processes. In the present study, we isolated and characterized EVs from the brains of rhesus macaques, both with and without simian immunodeficiency virus (SIV) induced central nervous system (CNS) disease. Small RNA sequencing revealed increased miR-21 levels in EVs from SIV encephalitic (SIVE) brains. In situ hybridization revealed increased miR-21 expression in neurons and macrophage/microglial cells/nodules during SIV induced CNS disease. In vitro culture of macrophages revealed that miR-21 is released into EVs and is neurotoxic when compared to EVs derived from miR-21-/- knockout animals. A mutation of the sequence within miR-21, predicted to bind TLR7, eliminates this neurotoxicity. Indeed miR-21 in EV activates TLR7 in a reporter cell line, and the neurotoxicity is dependent upon TLR7, as neurons isolated from TLR7-/- knockout mice are protected from neurotoxicity. Further, we show that EVs isolated from the brains of monkeys with SIV induced CNS disease activates TLR7 and were neurotoxic when compared to EVs from control animals. Finally, we show that EV-miR-21 induced neurotoxicity was unaffected by apoptosis inhibition but could be prevented by a necroptosis inhibitor, necrostatin-1, highlighting the actions of this pathway in a growing number of CNS disorders.


Assuntos
Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Receptor 7 Toll-Like/metabolismo , Animais , Western Blotting , Encéfalo/virologia , Imunofluorescência , Hibridização in Situ Fluorescente , Macaca mulatta , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/genética
14.
J Cell Mol Med ; 20(9): 1664-72, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27079462

RESUMO

Autophagy dysfunction in mouse atherosclerosis models has been associated with increased lipid accumulation, apoptosis and inflammation. Expression of cystatin C (CysC) is decreased in human atheroma, and CysC deficiency enhances atherosclerosis in mice. Here, we first investigated the association of autophagy and CysC expression levels with atheroma plaque severity in human atherosclerotic lesions. We found that autophagy proteins Atg5 and LC3ß in advanced human carotid atherosclerotic lesions are decreased, while markers of dysfunctional autophagy p62/SQSTM1 and ubiquitin are increased together with elevated levels of lipid accumulation and apoptosis. The expressions of LC3ß and Atg5 were positively associated with CysC expression. Second, we investigated whether CysC expression is involved in autophagy in atherosclerotic apoE-deficient mice, demonstrating that CysC deficiency (CysC(-/-) ) in these mice results in reduction of Atg5 and LC3ß levels and induction of apoptosis. Third, macrophages isolated from CysC(-/-) mice displayed increased levels of p62/SQSTM1 and higher sensitivity to 7-oxysterol-mediated lysosomal membrane destabilization and apoptosis. Finally, CysC treatment minimized oxysterol-mediated cellular lipid accumulation. We conclude that autophagy dysfunction is a characteristic of advanced human atherosclerotic lesions and is associated with reduced levels of CysC. The deficiency of CysC causes autophagy dysfunction and apoptosis in macrophages and apoE-deficient mice. The results indicate that CysC plays an important regulatory role in combating cell death via the autophagic pathway in atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Autofagia , Cistatina C/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Idoso , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular , Cistatina C/deficiência , Progressão da Doença , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Permeabilidade , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Proteínas Recombinantes/farmacologia
15.
J Neurosci ; 34(15): 5099-106, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24719089

RESUMO

Trisomy 21, or Down's syndrome (DS), is the most common genetic cause of intellectual disability. Altered neurotransmission in the brains of DS patients leads to hippocampus-dependent learning and memory deficiency. Although genetic mouse models have provided important insights into the genes and mechanisms responsible for DS-specific changes, the molecular mechanisms leading to memory deficits are not clear. We investigated whether the segmental trisomy model of DS, Ts[Rb(12.1716)]2Cje (Ts2), exhibits hippocampal glutamatergic transmission abnormalities and whether these alterations cause behavioral deficits. Behavioral assays demonstrated that Ts2 mice display a deficit in nest building behavior, a measure of hippocampus-dependent nonlearned behavior, as well as dysfunctional hippocampus-dependent spatial memory tested in the object-placement and the Y-maze spontaneous alternation tasks. Magnetic resonance spectra measured in the hippocampi revealed a significantly lower glutamate concentration in Ts2 as compared with normal disomic (2N) littermates. The glutamate deficit accompanied hippocampal NMDA receptor1 (NMDA-R1) mRNA and protein expression level downregulation in Ts2 compared with 2N mice. In concert with these alterations, paired-pulse analyses suggested enhanced synaptic inhibition and/or lack of facilitation in the dentate gyrus of Ts2 compared with 2N mice. Ts2 mice also exhibited disrupted synaptic plasticity in slice recordings of the hippocampal CA1 region. Collectively, these findings imply that deficits in glutamate and NMDA-R1 may be responsible for impairments in synaptic plasticity in the hippocampus associated with behavioral dysfunctions in Ts2 mice. Thus, these findings suggest that glutamatergic deficits have a significant role in causing intellectual disabilities in DS.


Assuntos
Síndrome de Down/metabolismo , Ácido Glutâmico/metabolismo , Potenciação de Longa Duração , Aprendizagem em Labirinto , Memória , Comportamento de Nidação , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Giro Denteado/metabolismo , Giro Denteado/fisiopatologia , Modelos Animais de Doenças , Síndrome de Down/fisiopatologia , Feminino , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia
16.
Cell Mol Life Sci ; 71(4): 673-82, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23995987

RESUMO

Computational approaches for detecting co-evolution in proteins allow for the identification of protein-protein interaction networks in different organisms and the assignment of function to under-explored proteins. The detection of co-variation of amino acids within or between proteins, moreover, allows for the discovery of residue-residue contacts and highlights functional residues that can affect the binding affinity, catalytic activity, or substrate specificity of a protein. To explore the functional impact of co-evolutionary changes in proteins, a combined experimental and computational approach must be recruited. Here, we review recent studies that apply computational and experimental tools to obtain novel insight into the structure, function, and evolution of proteins. Specifically, we describe the application of co-evolutionary analysis for predicting high-resolution three-dimensional structures of proteins. In addition, we describe computational approaches followed by experimental analysis for identifying specificity-determining residues in proteins. Finally, we discuss studies addressing the importance of such residues in terms of the functional divergence of proteins, allowing proteins to evolve new functions while avoiding crosstalk with existing cellular pathways or forming reproductive barriers and hence promoting speciation.


Assuntos
Evolução Molecular , Mapas de Interação de Proteínas , Proteínas/química , Proteínas/metabolismo , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Biologia Computacional/métodos , Especiação Genética , Humanos , Modelos Moleculares , Conformação Proteica , Proteínas/genética , Transdução de Sinais
17.
Mol Neurodegener ; 19(1): 34, 2024 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-38616258

RESUMO

BACKGROUND: Hypometabolism tied to mitochondrial dysfunction occurs in the aging brain and in neurodegenerative disorders, including in Alzheimer's disease, in Down syndrome, and in mouse models of these conditions. We have previously shown that mitovesicles, small extracellular vesicles (EVs) of mitochondrial origin, are altered in content and abundance in multiple brain conditions characterized by mitochondrial dysfunction. However, given their recent discovery, it is yet to be explored what mitovesicles regulate and modify, both under physiological conditions and in the diseased brain. In this study, we investigated the effects of mitovesicles on synaptic function, and the molecular players involved. METHODS: Hippocampal slices from wild-type mice were perfused with the three known types of EVs, mitovesicles, microvesicles, or exosomes, isolated from the brain of a mouse model of Down syndrome or of a diploid control and long-term potentiation (LTP) recorded. The role of the monoamine oxidases type B (MAO-B) and type A (MAO-A) in mitovesicle-driven LTP impairments was addressed by treatment of mitovesicles with the irreversible MAO inhibitors pargyline and clorgiline prior to perfusion of the hippocampal slices. RESULTS: Mitovesicles from the brain of the Down syndrome model reduced LTP within minutes of mitovesicle addition. Mitovesicles isolated from control brains did not trigger electrophysiological effects, nor did other types of brain EVs (microvesicles and exosomes) from any genotype tested. Depleting mitovesicles of their MAO-B, but not MAO-A, activity eliminated their ability to alter LTP. CONCLUSIONS: Mitovesicle impairment of LTP is a previously undescribed paracrine-like mechanism by which EVs modulate synaptic activity, demonstrating that mitovesicles are active participants in the propagation of cellular and functional homeostatic changes in the context of neurodegenerative disorders.


Assuntos
Doença de Alzheimer , Síndrome de Down , Doenças Mitocondriais , Humanos , Animais , Camundongos , Espaço Extracelular , Plasticidade Neuronal , Encéfalo , Modelos Animais de Doenças , Monoaminoxidase
18.
J Biol Chem ; 287(51): 43108-15, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23129776

RESUMO

In vitro studies have shown that neuronal cell cultures secrete exosomes containing amyloid-ß precursor protein (APP) and the APP-processing products, C-terminal fragments (CTFs) and amyloid-ß (Aß). We investigated the secretion of full-length APP (flAPP) and APP CTFs via the exosome secretory pathway in vivo. To this end, we developed a novel protocol designed to isolate exosomes secreted into mouse brain extracellular space. Exosomes with typical morphology were isolated from freshly removed mouse brains and from frozen mouse and human brain tissues, demonstrating that exosomes can be isolated from post-mortem tissue frozen for long periods of time. flAPP, APP CTFs, and enzymes that cleave both flAPP and APP CTFs were identified in brain exosomes. Although higher levels of both flAPP and APP CTFs were observed in exosomes isolated from the brains of transgenic mice overexpressing human APP (Tg2576) compared with wild-type control mice, there was no difference in the number of secreted brain exosomes. These data indicate that the levels of flAPP and APP CTFs associated with exosomes mirror the cellular levels of flAPP and APP CTFs. Interestingly, exosomes isolated from the brains of both Tg2576 and wild-type mice are enriched with APP CTFs relative to flAPP. Thus, we hypothesize that the exosome secretory pathway plays a pleiotropic role in the brain: exosome secretion is beneficial to the cell, acting as a specific releasing system of neurotoxic APP CTFs and Aß, but the secretion of exosomes enriched with APP CTFs, neurotoxic proteins that are also a source of secreted Aß, is harmful to the brain.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Exossomos/metabolismo , Espaço Extracelular/metabolismo , Via Secretória , Idoso , Animais , Exossomos/ultraestrutura , Feminino , Secções Congeladas , Humanos , Camundongos , Camundongos Transgênicos , Transporte Proteico
19.
J Extracell Vesicles ; 12(1): e12301, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36691887

RESUMO

Cocaine, an addictive psychostimulant, has a broad mechanism of action, including the induction of a wide range of alterations in brain metabolism and mitochondrial homeostasis. Our group recently identified a subpopulation of non-microvesicular, non-exosomal extracellular vesicles of mitochondrial origin (mitovesicles) and developed a method to isolate mitovesicles from brain parenchyma. We hypothesised that the generation and secretion of mitovesicles is affected by mitochondrial abnormalities induced by chronic cocaine exposure. Mitovesicles from the brain extracellular space of cocaine-administered mice were enlarged and more numerous when compared to controls, supporting a model in which mitovesicle biogenesis is enhanced in the presence of mitochondrial alterations. This interrelationship was confirmed in vitro. Moreover, cocaine affected mitovesicle protein composition, causing a functional alteration in mitovesicle ATP production capacity. These data suggest that mitovesicles are previously unidentified players in the biology of cocaine addiction and that target therapies to fine-tune brain mitovesicle functionality may be beneficial to mitigate the effects of chronic cocaine exposure.


Assuntos
Cocaína , Vesículas Extracelulares , Camundongos , Animais , Cocaína/metabolismo , Cocaína/farmacologia , Vesículas Extracelulares/metabolismo , Encéfalo/metabolismo , Mitocôndrias/metabolismo , Biologia
20.
Ann N Y Acad Sci ; 1523(1): 24-37, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36961472

RESUMO

Extracellular vesicles (EVs) are small, lipid-bilayer-bound particles released by cells that can contain important bioactive molecules, including lipids, RNAs, and proteins. Once released in the extracellular environment, EVs can act as messengers locally as well as to distant tissues to coordinate tissue homeostasis and systemic responses. There is a growing interest in not only understanding the physiology of EVs as signaling particles but also leveraging them as minimally invasive diagnostic and prognostic biomarkers (e.g., they can be found in biofluids) and drug-delivery vehicles. On October 30-November 2, 2022, researchers in the EV field convened for the Keystone symposium "Exosomes, Microvesicles, and Other Extracellular Vesicles" to discuss developing standardized language and methodology, new data on the basic biology of EVs and potential clinical utility, as well as novel technologies to isolate and characterize EVs.


Assuntos
Micropartículas Derivadas de Células , Exossomos , Vesículas Extracelulares , Humanos , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Micropartículas Derivadas de Células/metabolismo , RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA