Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 14: 1308647, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38143917

RESUMO

Introduction: Teaching methodologies promoting active learning result in higher-order knowledge application, a desirable outcome in health disciplines like Physiology. Flipped-classroom (FC) promotes active learning and engagement in the classroom. Although specialized research keeps accumulating, the advantages of FC for improving academic outcome and ultimately patient care remain controversial and open to further analysis. Objective: This study evaluates the benefits of applying FC to the Neurophysiology module of a Human Physiology course. Methods:We compare final grades of students exposed to standard lecturing (five-years) vs. FC (six-years), and study the FC impact on student motivation, study time and rewards. Differing from conventional FC, we performed no pre-class/in-class assessments, relying on the students' internal motivation to experience our FC model. A printed student workbook was designed as pre-class material for each session. Reading times respect the expected daily study time of students in our system. Results and discussion: Concerning academic performance, our long-term study reports a significant increase in average scores for FC groups. Overall, students get better scores in multiple choice tests than in problem-solving questions. A more detailed analysis uncovers that our FC model helps students to obtain better scores, reducing variability in performance due to assessment methods. Based on our open-ended survey questions, most students rate the FC environment and in-class activities positively and perceive a positive effect of FC on teachers' performance. An objective automatic Sentiment analysis of open-ended answers reveals that FC is positively appreciated by students, associating positive perceptions to their understanding of physiological concepts, and negative evaluations to their time management.

2.
Front Cell Neurosci ; 17: 1112930, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36779011

RESUMO

Microglial cells are recognized as very dynamic brain cells, screening the environment and sensitive to signals from all other cell types in health and disease. Apolipoprotein D (ApoD), a lipid-binding protein of the Lipocalin family, is required for nervous system optimal function and proper development and maintenance of key neural structures. ApoD has a cell and state-dependent expression in the healthy nervous system, and increases its expression upon aging, damage or neurodegeneration. An extensive overlap exists between processes where ApoD is involved and those where microglia have an active role. However, no study has analyzed the role of ApoD in microglial responses. In this work, we test the hypothesis that ApoD, as an extracellular signal, participates in the intercellular crosstalk sensed by microglia and impacts their responses upon physiological aging or damaging conditions. We find that a significant proportion of ApoD-dependent aging transcriptome are microglia-specific genes, and show that lack of ApoD in vivo dysregulates microglial density in mouse hippocampus in an age-dependent manner. Murine BV2 and primary microglia do not express ApoD, but it can be internalized and targeted to lysosomes, where unlike other cell types it is transiently present. Cytokine secretion profiles and myelin phagocytosis reveal that ApoD has both long-term pre-conditioning effects on microglia as well as acute effects on these microglial immune functions, without significant modification of cell survival. ApoD-triggered cytokine signatures are stimuli (paraquat vs. Aß oligomers) and sex-dependent. Acute exposure to ApoD induces microglia to switch from their resting state to a secretory and less phagocytic phenotype, while long-term absence of ApoD leads to attenuated cytokine induction and increased myelin uptake, supporting a role for ApoD as priming or immune training factor. This knowledge should help to advance our understanding of the complex responses of microglia during aging and neurodegeneration, where signals received along our lifespan are combined with damage-triggered acute signals, conditioning both beneficial roles and limitations of microglial functions.

3.
Mol Neurobiol ; 59(7): 4015-4029, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35460054

RESUMO

Accumulated evidence points to the lipocalin apolipoprotein D (ApoD), one of the few genes consistently upregulated upon brain ageing and neurodegeneration, as an endogenous controller of the redox state of cellular and extracellular lipid structures. This biochemical function has downstream consequences as apparently varied as control of glycocalyx and myelin compaction, cell viability upon oxidative stress or modulation of signalling pathways. In spite of this knowledge, it is still unclear if ApoD function requires canonical receptor-mediated transductions systems. This work aims to examine ApoD-cell membrane interaction and its dependence on a proposed ApoD receptor, Basigin. Whole and fractionated membrane preparations from the brain, primary astrocytes, glial and neuronal cell lines, reveal ApoD as a very specific component of particular subtypes of detergent-resistant microdomains (DRMs). ApoD interacts in vitro with neuronal membranes and is stably associated with astrocytic membranes. ApoD associates with DRMs with specific buoyancy properties that co-fractionate with plasma or late-endosome-lysosome markers. A mass spectrometry analysis reveals that these Triton X-114 DRMs contain both plasma membrane and endosomal-lysosomal compartment lipid raft proteins. ApoD-DRM association is maintained under metabolic and acute oxidative stress conditions. However, ApoD-membrane interaction, its internalization and its lipid-antioxidant function do not require the presence of Basigin. This work supports a stable association of ApoD with membranes, independent of Basigin, and provides the basis to fully understand ApoD antioxidant neuroprotective mechanism as a mechanism taking place in specific membrane subdomains.


Assuntos
Basigina , Detergentes , Antioxidantes , Apolipoproteínas D/metabolismo , Lipocalinas , Microdomínios da Membrana/metabolismo
4.
Cells ; 11(2)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35053342

RESUMO

The insulin-degrading enzyme (IDE) is a zinc-dependent metalloendopeptidase that belongs to the M16A metalloprotease family. IDE is markedly expressed in the brain, where it is particularly relevant due to its in vitro amyloid beta (Aß)-degrading activity. The subcellular localization of IDE, a paramount aspect to understand how this enzyme can perform its proteolytic functions in vivo, remains highly controversial. In this work, we addressed IDE subcellular localization from an evolutionary perspective. Phylogenetic analyses based on protein sequence and gene and protein structure were performed. An in silico analysis of IDE signal peptide suggests an evolutionary shift in IDE exportation at the prokaryote/eukaryote divide. Subcellular localization experiments in microglia revealed that IDE is mostly cytosolic. Furthermore, IDE associates to membranes by their cytoplasmatic side and further partitions between raft and non-raft domains. When stimulated, microglia change into a secretory active state, produces numerous multivesicular bodies and IDE associates with their membranes. The subsequent inward budding of such membranes internalizes IDE in intraluminal vesicles, which later allows IDE to be exported outside the cells in small extracellular vesicles. We further demonstrate that such an IDE exportation mechanism is regulated by stimuli relevant for microglia in physiological conditions and upon aging and neurodegeneration.


Assuntos
Evolução Molecular , Insulisina/metabolismo , Microglia/enzimologia , Animais , Linhagem Celular , Células Cultivadas , Sequência Conservada , Citosol/metabolismo , Vesículas Extracelulares/metabolismo , Insulisina/ultraestrutura , Microdomínios da Membrana/metabolismo , Metaloendopeptidases/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/ultraestrutura , Corpos Multivesiculares/metabolismo , Filogenia , Frações Subcelulares/metabolismo
5.
Front Physiol ; 12: 738991, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34690812

RESUMO

Apolipoprotein D is a chordate gene early originated in the Lipocalin protein family. Among other features, regulation of its expression in a wide variety of disease conditions in humans, as apparently unrelated as neurodegeneration or breast cancer, have called for attention on this gene. Also, its presence in different tissues, from blood to brain, and different subcellular locations, from HDL lipoparticles to the interior of lysosomes or the surface of extracellular vesicles, poses an interesting challenge in deciphering its physiological function: Is ApoD a moonlighting protein, serving different roles in different cellular compartments, tissues, or organisms? Or does it have a unique biochemical mechanism of action that accounts for such apparently diverse roles in different physiological situations? To answer these questions, we have performed a systematic review of all primary publications where ApoD properties have been investigated in chordates. We conclude that ApoD ligand binding in the Lipocalin pocket, combined with an antioxidant activity performed at the rim of the pocket are properties sufficient to explain ApoD association with different lipid-based structures, where its physiological function is better described as lipid-management than by long-range lipid-transport. Controlling the redox state of these lipid structures in particular subcellular locations or extracellular structures, ApoD is able to modulate an enormous array of apparently diverse processes in the organism, both in health and disease. The new picture emerging from these data should help to put the physiological role of ApoD in new contexts and to inspire well-focused future research.

6.
Front Physiol ; 12: 718983, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34497539

RESUMO

The protein family of Lipocalins is ubiquitously present throughout the tree of life, with the exception of the phylum Archaea. Phylogenetic relationships of chordate Lipocalins have been proposed in the past based on protein sequence similarities, but their highly divergent primary structures and a shortage of experimental annotations in genome projects have precluded a well-supported hypothesis for their evolution. In this work we propose a novel topology for the phylogenetic tree of chordate Lipocalins, inferred from multiple amino acid sequence alignments. Sixteen jawed vertebrates with fair coverage by genomic sequencing were compared. The selected species span an evolutionary range of ∼400 million years, allowing for a balanced representation of all major vertebrate clades. A consensus phylogenetic tree is proposed following a comparison of sequence-based maximum-likelihood trees and protein structure dendrograms. This new phylogeny suggests an APOD-like common ancestor in early chordates, which gave rise, via whole-genome or tandem duplications, to the six Lipocalins currently present in fish (APOD, RBP4, PTGDS, AMBP, C8G, and APOM). Further gene duplications of APOM and PTGDS resulted in the altogether 15 Lipocalins found in contemporary mammals. Insights into the functional impact of relevant amino acid residues in early diverging Lipocalins are also discussed. These results should foster the experimental exploration of novel functions alongside the identification of new members of the Lipocalin family.

7.
Neurobiol Dis ; 144: 105046, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32798728

RESUMO

Lysosomal Storage Diseases (LSD) are genetic diseases causing systemic and nervous system dysfunction. The glia-derived lipid binding protein Apolipoprotein D (ApoD) is required for lysosomal functional integrity in glial and neuronal cells, ensuring cell survival upon oxidative stress or injury. Here we test whether ApoD counteracts the pathogenic consequences of a LSD, Niemann Pick-type-A disease (NPA), where mutations in the acid sphingomyelinase gene result in sphingomyelin accumulation, lysosomal permeabilization and early-onset neurodegeneration. We performed a multivariable analysis of behavioral, cellular and molecular outputs in 12 and 24 week-old male and female NPA model mice, combined with ApoD loss-of-function mutation. Lack of ApoD in NPA mice accelerates cerebellar-dependent motor deficits, enhancing loss of Purkinje neurons. We studied ApoD expression in brain sections from a NPA patient and age-matched control, and the functional consequences of ApoD supplementation in primary human fibroblasts from two independent NPA patients and two control subjects. Cell viability, lipid peroxidation, and lysosomal functional integrity (pH, Cathepsin B activity, Galectin-3 exclusion) were examined. ApoD is endogenously overexpressed in NPA patients and NPA mouse brains and targeted to lysosomes of NPA patient cells, including Purkinje neurons and cultured fibroblasts. The accelerated lysosomal targeting of ApoD by oxidative stress is hindered in NPA fibroblasts, contributing to NPA lysosomes vulnerability. Exogenously added ApoD reduces NPA-prompted lysosomal permeabilization and alkalinization, reverts lipid peroxides accumulation, and significantly increases NPA cell survival. ApoD administered simultaneously to sphingomyelin overload results in complete rescue of cell survival. Our results reveal that ApoD protection of lysosomal integrity counteracts NPA pathology. ApoD supplementation could significantly delay not only the progression of NPA disease, but also of other LSDs through its beneficial effects in lysosomal functional maintenance.


Assuntos
Apolipoproteínas D/genética , Lisossomos/metabolismo , Atividade Motora/genética , Doença de Niemann-Pick Tipo A/fisiopatologia , Animais , Apolipoproteínas D/farmacologia , Comportamento Animal , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Pré-Escolar , Progressão da Doença , Humanos , Camundongos , Camundongos Knockout , Doença de Niemann-Pick Tipo A/genética , Doença de Niemann-Pick Tipo A/metabolismo , Teste de Campo Aberto , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Paraquat , Permeabilidade , Teste de Desempenho do Rota-Rod , Esfingomielina Fosfodiesterase/genética
8.
Mar Drugs ; 18(8)2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32708004

RESUMO

Neurodegenerative diseases are age-related disorders caused by progressive neuronal death in different regions of the nervous system. Neuroinflammation, modulated by glial cells, is a crucial event during the neurodegenerative process; consequently, there is an urgency to find new therapeutic products with anti-glioinflammatory properties. Five new furanocembranolides (1-5), along with leptolide, were isolated from two different extracts of Leptogorgia sp., and compound 6 was obtained from chemical transformation of leptolide. Their structures were determined based on spectroscopic evidence. These seven furanocembranolides were screened in vitro by measuring their ability to modulate interleukin-1ß (IL-1ß) production by microglial BV2 cells after LPS (lipopolysaccharide) stimulation. Leptolide and compounds 3, 4 and 6 exhibited clear anti-inflammatory effects on microglial cells, while compound 2 presented a pro-inflammatory outcome. The in vitro results prompted us to assess anti-glioinflammatory effects of leptolide in vivo in a high-fat diet-induced obese mouse model. Interestingly, leptolide treatment ameliorated both microgliosis and astrogliosis in this animal model. Taken together, our results reveal a promising direct biological effect of furanocembranolides on microglial cells as bioactive anti-inflammatory molecules. Among them, leptolide provides us a feasible therapeutic approach to treat neuroinflammation concomitant with metabolic impairment.


Assuntos
Anti-Inflamatórios/farmacologia , Encéfalo/efeitos dos fármacos , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Diterpenos/farmacologia , Furanos/farmacologia , Gliose/tratamento farmacológico , Resistência à Insulina , Microglia/efeitos dos fármacos , Obesidade/complicações , Animais , Antozoários/química , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Encéfalo/metabolismo , Encéfalo/patologia , Hidrocarbonetos Aromáticos com Pontes/química , Hidrocarbonetos Aromáticos com Pontes/isolamento & purificação , Linhagem Celular , Dieta Hiperlipídica , Diterpenos/química , Diterpenos/isolamento & purificação , Furanos/química , Furanos/isolamento & purificação , Gliose/etiologia , Gliose/metabolismo , Gliose/patologia , Interleucina-1beta/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Estrutura Molecular , Obesidade/metabolismo , Relação Estrutura-Atividade
9.
Front Neurosci ; 14: 516, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32581679

RESUMO

The fruit fly compound eye is a premier experimental system for modeling human neurodegenerative diseases. The disruption of the retinal geometry has been historically assessed using time-consuming and poorly reliable techniques such as histology or pseudopupil manual counting. Recent semiautomated quantification approaches rely either on manual region-of-interest delimitation or engineered features to estimate the extent of degeneration. This work presents a fully automated classification pipeline of bright-field images based on orientated gradient descriptors and machine learning techniques. An initial region-of-interest extraction is performed, applying morphological kernels and Euclidean distance-to-centroid thresholding. Image classification algorithms are trained on these regions (support vector machine, decision trees, random forest, and convolutional neural network), and their performance is evaluated on independent, unseen datasets. The combinations of oriented gradient + gaussian kernel Support Vector Machine [0.97 accuracy and 0.98 area under the curve (AUC)] and fine-tuned pre-trained convolutional neural network (0.98 accuracy and 0.99 AUC) yielded the best results overall. The proposed method provides a robust quantification framework that can be generalized to address the loss of regularity in biological patterns similar to the Drosophila eye surface and speeds up the processing of large sample batches.

10.
PLoS One ; 15(6): e0234857, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32559215

RESUMO

The Lipocalin Apolipoprotein D (ApoD) is one of the few genes consistently overexpressed in the aging brain, and in most neurodegenerative and psychiatric diseases. Its functions include metabolism regulation, myelin management, neuroprotection, and longevity regulation. Knowledge of endogenous regulatory mechanisms controlling brain disease-triggered ApoD expression is relevant if we want to boost pharmacologically its neuroprotecting potential. In addition to classical transcriptional control, Lipocalins have a remarkable variability in mRNA 5'UTR-dependent translation efficiency. Using bioinformatic analyses, we uncover strong selective pressures preserving ApoD 5'UTR properties, indicating unexpected functional conservation. PCR amplifications demonstrate the production of five 5'UTR variants (A-E) in mouse ApoD, with diverse expression levels across tissues and developmental stages. Importantly, Variant E is specifically expressed in the oxidative stress-challenged brain. Predictive analyses of 5'UTR secondary structures and enrichment in elements restraining translation, point to Variant E as a tight regulator of ApoD expression. We find two genomic regions conserved in human and mouse ApoD: a canonical (α) promoter region and a previously unknown region upstream of Variant E that could function as an alternative mouse promoter (ß). Luciferase assays demonstrate that both α and ß promoter regions can drive expression in cultured mouse astrocytes, and that Promoter ß activity responds proportionally to incremental doses of the oxidative stress generator Paraquat. We postulate that Promoter ß works in association with Variant E 5'UTR as a regulatory tandem that organizes ApoD gene expression in the nervous system in response to oxidative stress, the most common factor in aging and neurodegeneration.


Assuntos
Regiões 5' não Traduzidas , Apolipoproteínas D/genética , Apolipoproteínas E/genética , Regiões Promotoras Genéticas , Animais , Apolipoproteínas D/metabolismo , Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Herbicidas/toxicidade , Lipocalinas/genética , Lipocalinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Estresse Oxidativo , Paraquat/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Front Neurol ; 10: 1152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31787919

RESUMO

A proper lipid management is paramount for a healthy brain. Lipid homeostasis alterations are known to be causative or risk factors for many neurodegenerative diseases, or key elements in the recovery from nervous system injuries of different etiology. In addition to lipid biogenesis and catabolism, non-enzymatic lipid-binding proteins play an important role in brain function and maintenance through aging. Among these types of lipoproteins, apolipoprotein E has received much attention due to the relationship of particular alleles of its gene with the risk and progression of Alzheimer's disease. However, other lipid-binding proteins whose role in lipid homeostasis and control are less known need to be brought to the attention of both researchers and clinicians. The aim of this review is to cover the knowledge of lipid-managing proteins in the brain, with particular attention to new candidates to be relevant for brain function and health.

12.
PLoS One ; 14(3): e0213206, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30840684

RESUMO

The Lipocalin family is a group of homologous proteins characterized by its big array of functional capabilities. As extracellular proteins, they can bind small hydrophobic ligands through a well-conserved ß-barrel folding. Lipocalins evolutionary history sprawls across many different taxa and shows great divergence even within chordates. This variability is also found in their heterogeneous tissue expression pattern. Although a handful of promoter regions have been previously described, studies on UTR regulatory roles in Lipocalin gene expression are scarce. Here we report a comprehensive bioinformatic analysis showing that complex post-transcriptional regulation exists in Lipocalin genes, as suggested by the presence of alternative UTRs with substantial sequence conservation in mammals, alongside a high diversity of transcription start sites and alternative promoters. Strong selective pressure could have operated upon Lipocalins UTRs, leading to an enrichment in particular sequence motifs that limit the choice of secondary structures. Mapping these regulatory features to the expression pattern of early and late diverging Lipocalins suggests that UTRs represent an additional phylogenetic signal, which may help to uncover how functional pleiotropy originated within the Lipocalin family.


Assuntos
Simulação por Computador , Evolução Molecular , Lipocalinas/genética , Proteínas/metabolismo , Processamento Pós-Transcricional do RNA , Regiões não Traduzidas/genética , Animais , Biologia Computacional , Humanos , Lipocalinas/metabolismo , Mamíferos , Filogenia , Proteínas/genética , Sítio de Iniciação de Transcrição
13.
Acta Histochem ; 120(3): 179-186, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29395318

RESUMO

The MTT assay was the first widely accepted method to assess cytotoxicity and cell viability. However, there is controversy on whether this indicator is a useful tool. In this work we intend to expand the interpretability of the MTT study by its combination with widely used cellular biology techniques. We propose complementary approaches to the colorimetric assay, based on the use of measurements in three different settings: confocal microscopy, multi-well plate assay and flow cytometry. Using confocal microscopy, we confirmed that MTT uptake and reduction by cells is a time-dependent process, and that formazan accumulates in round-shaped organelles. Quantitative measurements with a multi-well fluorimeter combined with nuclear staining result in a useful method, yielding a ratio between formazan production and cell number that informs about the average cell metabolic state. We also found that flow cytometry is a suitable technique to measure MTT reduction in large cell populations. When assaying the effect of an oxidizing agent such as paraquat (PQ), this approach allows for the distinction of subpopulations of cells with different reducing power. Finally, we prove that it is feasible to monitor MTT reduction in an in vivo model, the Drosophila larvae, without affecting its survival rate. Formazan accumulates exclusively in the larval fat body, confirming its lipid solubility. The methods explored in this work expand the MTT potential as a useful tool to provide information of the physiological state of cells and organisms.


Assuntos
Formazans , Larva/efeitos dos fármacos , Paraquat/farmacologia , Sais de Tetrazólio , Animais , Bioensaio , Contagem de Células , Drosophila/efeitos dos fármacos , Corpo Adiposo/efeitos dos fármacos , Citometria de Fluxo , Formazans/química , Células HeLa , Humanos , Lipídeos/farmacocinética , Microscopia Confocal , Oxirredução , Paraquat/farmacocinética , Solubilidade , Sais de Tetrazólio/química , Fatores de Tempo
14.
Front Cell Neurosci ; 12: 526, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687015

RESUMO

Extracellular vesicle (EV)-mediated glia-to-neuron communication has been recognized in a growing number of physiological and pathological situations. They transport complex sets of molecules that can be beneficial or detrimental for the receiving cell. As in other areas of biology, their analysis is revolutionizing the field of neuroscience, since fundamental signaling processes are being re-evaluated, and applications for neurodegenerative disease therapies have emerged. Using human astrocytic and differentiated neuronal cell lines, we demonstrate that a classical neuroprotective protein, Apolipoprotein D (ApoD), expressed by glial cells and known to promote functional integrity and survival of neurons, is exclusively transported by EVs from astrocytes to neurons, where it gets internalized. Indeed, we demonstrate that conditioned media derived from ApoD-knock-out (KO) astrocytes exert only a partial autocrine protection from oxidative stress (OS) challenges, and that EVs are required for ApoD-positive astrocytic cell line derived medium to exert full neuroprotection. When subfractionation of EVs is performed, ApoD is revealed as a very specific marker of the exosome-containing fractions. These discoveries help us reframe our understanding of the neuroprotective role of this lipid binding protein and open up new research avenues to explore the use of systemically administered ApoD-loaded exosomes that can cross the blood-brain barrier to treat neurodegenerative diseases.

15.
Glia ; 66(3): 670-687, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29222871

RESUMO

To compact the extracellular sides of myelin, an important transition must take place: from membrane sliding, while building the wraps, to membrane adhesion and water exclusion. Removal of the negatively charged glycocalyx becomes the limiting factor in such transition. What is required to initiate this membrane-zipping process? Knocking-out the Lipocalin Apolipoprotein D (ApoD), essential for lysosomal functional integrity in glial cells, results in a specific defect in myelin extracellular leaflet compaction in peripheral and central nervous system, which results in reduced conduction velocity and suboptimal behavioral outputs: motor learning is compromised. Myelination initiation, growth, intracellular leaflet compaction, myelin thickness or internodal length remain unaltered. Lack of ApoD specifically modifies Plp and P0 protein expression, but not Mbp or Mag. Late in myelin maturation period, ApoD affects lipogenic and growth-related, but not stress-responsive, signaling pathways. Without ApoD, the sialylated glycocalyx is maintained and ganglioside content remains high. In peripheral nervous system, Neu3 membrane sialidase and lysosomal Neu1 are coordinately expressed with ApoD in subsets of Schwann cells. ApoD-KO myelin becomes depleted of Neu3 and enriched in Fyn, a kinase with pivotal roles in transducing axon-derived signals into myelin properties. In the absence of ApoD, partial permeabilization of lysosomes alters Neu1 location as well. Exogenous ApoD rescues ApoD-KO hypersialylated glycocalyx in astrocytes, demonstrating that ApoD is necessary and sufficient to control glycocalyx composition in glial cells. By ensuring lysosomal functional integrity and adequate subcellular location of effector and regulatory proteins, ApoD guarantees the glycolipid recycling and glycocalyx removal required to complete myelin compaction.


Assuntos
Apolipoproteínas D/metabolismo , Glicocálix/metabolismo , Lisossomos/metabolismo , Bainha de Mielina/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Apolipoproteínas D/administração & dosagem , Apolipoproteínas D/genética , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Escherichia coli , Espaço Extracelular/metabolismo , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Mucolipidoses/metabolismo , Neuraminidase/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Nervo Isquiático/citologia , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo
16.
PLoS Genet ; 13(2): e1006603, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28182653

RESUMO

Environmental insults such as oxidative stress can damage cell membranes. Lysosomes are particularly sensitive to membrane permeabilization since their function depends on intraluminal acidic pH and requires stable membrane-dependent proton gradients. Among the catalog of oxidative stress-responsive genes is the Lipocalin Apolipoprotein D (ApoD), an extracellular lipid binding protein endowed with antioxidant capacity. Within the nervous system, cell types in the defense frontline, such as astrocytes, secrete ApoD to help neurons cope with the challenge. The protecting role of ApoD is known from cellular to organism level, and many of its downstream effects, including optimization of autophagy upon neurodegeneration, have been described. However, we still cannot assign a cellular mechanism to ApoD gene that explains how this protection is accomplished. Here we perform a comprehensive analysis of ApoD intracellular traffic and demonstrate its role in lysosomal pH homeostasis upon paraquat-induced oxidative stress. By combining single-lysosome in vivo pH measurements with immunodetection, we demonstrate that ApoD is endocytosed and targeted to a subset of vulnerable lysosomes in a stress-dependent manner. ApoD is functionally stable in this acidic environment, and its presence is sufficient and necessary for lysosomes to recover from oxidation-induced alkalinization, both in astrocytes and neurons. This function is accomplished by preventing lysosomal membrane permeabilization. Two lysosomal-dependent biological processes, myelin phagocytosis by astrocytes and optimization of neurodegeneration-triggered autophagy in a Drosophila in vivo model, require ApoD-related Lipocalins. Our results uncover a previously unknown biological function of ApoD, member of the finely regulated and evolutionary conserved gene family of extracellular Lipocalins. They set a lipoprotein-mediated regulation of lysosomal membrane integrity as a new mechanism at the hub of many cellular functions, critical for the outcome of a wide variety of neurodegenerative diseases. These results open therapeutic opportunities by providing a route of entry and a repair mechanism for lysosomes in pathological situations.


Assuntos
Astrócitos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Apolipoproteínas D/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/ultraestrutura , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Células Cultivadas , Drosophila , Células HEK293 , Herbicidas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting , Lipocalinas/farmacologia , Lisossomos/química , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Neurônios/efeitos dos fármacos , Paraquat/farmacologia , Fagossomos/metabolismo
17.
Exp Gerontol ; 67: 19-47, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25868396

RESUMO

A detailed knowledge of the mechanisms underlying brain aging is fundamental to understand its functional decline and the baseline upon which brain pathologies superimpose. Endogenous protective mechanisms must contribute to the adaptability and plasticity still present in the healthy aged brain. Apolipoprotein D (ApoD) is one of the few genes with a consistent and evolutionarily conserved up-regulation in the aged brain. ApoD protecting roles upon stress or injury are well known, but a study of the effects of ApoD expression in the normal aging process is still missing. Using an ApoD-knockout mouse we analyze the effects of ApoD on factors contributing to the functional maintenance of the aged brain. We focused our cellular and molecular analyses in the cortex and hippocampus at an age representing the onset of senescence where mortality risks are below 25%, avoiding bias towards long-lived animals. Lack of ApoD causes a prematurely aged brain without altering lifespan. Age-dependent hyperkinesia and memory deficits are accompanied by differential molecular effects in the cortex and hippocampus. Transcriptome analyses reveal distinct effects of ApoD loss on the molecular age-dependent patterns of the cortex and hippocampus, with different cell-type contributions to age-regulated gene expression. Markers of glial reactivity, proteostasis, and oxidative and inflammatory damage reveal early signs of aging and enhanced brain deterioration in the ApoD-knockout brain. The lack of ApoD results in an age-enhanced significant reduction in neuronal calcium-dependent functionality markers and signs of early reduction of neuronal numbers in the cortex, thus impinging upon parameters clearly differentiating neurodegenerative conditions from healthy brain aging. Our data support the hypothesis that the physiological increased brain expression of ApoD represents a homeostatic anti-aging mechanism.


Assuntos
Envelhecimento/metabolismo , Apolipoproteínas D/fisiologia , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Senilidade Prematura/genética , Senilidade Prematura/metabolismo , Senilidade Prematura/patologia , Animais , Apolipoproteínas D/deficiência , Apolipoproteínas D/genética , Comportamento Animal , Córtex Cerebral/patologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Feminino , Regulação da Expressão Gênica/fisiologia , Hipocampo/patologia , Masculino , Camundongos Knockout , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Transcriptoma
18.
Mol Neurodegener ; 10: 9, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25887846

RESUMO

The fruitfly compound eye has been broadly used as a model for neurodegenerative diseases. Classical quantitative techniques to estimate the degeneration level of an eye under certain experimental conditions rely either on time consuming histological techniques to measure retinal thickness, or pseudopupil visualization and manual counting. Alternatively, visual examination of the eye surface appearance gives only a qualitative approximation provided the observer is well-trained. Therefore, there is a need for a simplified and standardized analysis of fruitfly eye degeneration extent for both routine laboratory use and for automated high-throughput analysis. We have designed the freely available ImageJ plugin FLEYE, a novel and user-friendly method for quantitative unbiased evaluation of neurodegeneration levels based on the acquisition of fly eye surface pictures. The incorporation of automated image analysis tools and a classification algorithm sustained on a built-in statistical model allow the user to quickly analyze large sample size data with reliability and robustness. Pharmacological screenings or genetic studies using the Drosophila retina as a model system may benefit from our method, because it can be easily implemented in a fully automated environment. In addition, FLEYE can be trained to optimize the image detection capabilities, resulting in a versatile approach to evaluate the pattern regularity of other biological or non-biological samples and their experimental or pathological disruption.


Assuntos
Comportamento Animal/fisiologia , Retina/patologia , Animais , Modelos Animais de Doenças , Drosophila melanogaster , Processamento de Imagem Assistida por Computador , Modelos Biológicos , Reprodutibilidade dos Testes
19.
Mol Neurodegener ; 10: 11, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25888134

RESUMO

BACKGROUND: A diverse set of neurodegenerative disorders are caused by abnormal extensions of polyglutamine (poly-Q) stretches in various, functionally unrelated proteins. A common feature of these diseases is altered proteostasis. Autophagy induction is part of the endogenous response to poly-Q protein expression. However, if autophagy is not resolved properly, clearance of toxic proteins or aggregates cannot occur effectively. Likewise, excessive autophagy induction can cause autophagic stress and neurodegeneration. The Lipocalins ApoD, Glial Lazarillo (GLaz) and Neural Lazarillo (NLaz) are neuroprotectors upon oxidative stress or aging. In this work we test whether these Lipocalins also protect against poly-Q-triggered deterioration of protein quality control systems. RESULTS: Using a Drosophila retinal degeneration model of Type-1 Spinocerebellar Ataxia (SCA1) combined with genetic manipulation of NLaz and GLaz expression, we demonstrate that both Lipocalins protect against SCA1 neurodegeneration. They are part of the endogenous transcriptional response to SCA1, and their effect is non-additive, suggesting participation in a similar mechanism. GLaz beneficial effects persist throughout aging, and appears when expressed by degenerating neurons or by retinal support and glial cells. GLaz gain-of-function reduces cell death and the extent of ubiquitinated proteins accumulation, and decreases the expression of Atg8a/LC3, p62 mRNA and protein levels, and GstS1 induction. Over-expression of GLaz is able to reduce p62 and ubiquitinated proteins levels when rapamycin-dependent and SCA1-dependent inductions of autophagy are combined. In the absence of neurodegeneration, GLaz loss-of-function increases Atg8a/LC3 mRNA and p62 protein levels without altering p62 mRNA levels. Knocking-down autophagy, by interfering with Atg8a or p62 expression or by expressing dominant-negative Atg1/ULK1 or Atg4a transgenes, rescues SCA1-dependent neurodegeneration in a similar extent to the protective effect of GLaz. Further GLaz-dependent improvement is concealed. CONCLUSIONS: This work shows for the first time that a Lipocalin rescues neurons from pathogenic SCA1 degeneration by optimizing clearance of aggregation-prone proteins. GLaz modulates key autophagy genes and lipid-peroxide clearance responsive genes. Down-regulation of selective autophagy causes similar and non-additive rescuing effects. These data suggest that SCA1 neurodegeneration concurs with autophagic stress, and places Lazarillo-related Lipocalins as valuable players in the endogenous protection against the two major contributors to aging and neurodegeneration: ROS-dependent damage and proteostasis deterioration.


Assuntos
Autofagia , Lipocalinas/metabolismo , Ataxias Espinocerebelares/patologia , Animais , Autofagia/genética , Autofagia/fisiologia , Morte Celular/genética , Morte Celular/fisiologia , Regulação para Baixo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Ataxias Espinocerebelares/genética , Tempo
20.
Neurobiol Aging ; 36(5): 1820-33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25784209

RESUMO

Apolipoprotein D (apoD) is expressed in the brain and levels are increased in affected brain regions in Alzheimer's disease (AD). The role that apoD may play in regulating AD pathology has not been addressed. Here, we crossed both apoD-null mice and Thy-1 human apoD transgenic mice with APP-PS1 amyloidogenic AD mice. Loss of apoD resulted in a nearly 2-fold increase in hippocampal amyloid plaque load, as assessed by immunohistochemical staining. Conversely, transgenic expression of neuronal apoD reduced hippocampal plaque load by approximately 35%. This latter finding was associated with a 60% decrease in amyloid ß 1-40 peptide levels, and a 34% decrease in insoluble amyloid ß 1-42 peptide. Assessment of ß-site amyloid precursor protein cleaving enzyme-1 (BACE1) levels and proteolytic products of amyloid precursor protein and neuregulin-1 point toward a possible association of altered BACE1 activity in association with altered apoD levels. In conclusion, the current studies provide clear evidence that apoD regulates amyloid plaque pathology in a mouse model of AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Apolipoproteínas D/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Camundongos Transgênicos , Neuregulina-1/metabolismo , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA