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











Base de dados
Intervalo de ano de publicação
1.
Cells ; 11(21)2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36359774

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder associated with neuron-glia dysfunction and dysregulated miRNAs. We previously reported upregulated miR-124/miR-21 in AD neurons and their exosomes. However, their glial distribution, phenotypic alterations and exosomal spread are scarcely documented. Here, we show glial cell activation and miR-21 overexpression in mouse organotypic hippocampal slices transplanted with SH-SY5Y cells expressing the human APP695 Swedish mutation. The upregulation of miR-21 only in the CSF from a small series of mild cognitive impairment (MCI) AD patients, but not in non-AD MCI individuals, supports its discriminatory potential. Microglia, neurons, and astrocytes differentiated from the same induced pluripotent stem cells from PSEN1ΔE9 AD patients all showed miR-21 elevation. In AD neurons, miR-124/miR-21 overexpression was recapitulated in their exosomes. In AD microglia, the upregulation of iNOS and miR-21/miR-146a supports their activation. AD astrocytes manifested a restrained inflammatory profile, with high miR-21 but low miR-155 and depleted exosomal miRNAs. Their immunostimulation with C1q + IL-1α + TNF-α induced morphological alterations and increased S100B, inflammatory transcripts, sAPPß, cytokine release and exosomal miR-21. PPARα, a target of miR-21, was found to be repressed in all models, except in neurons, likely due to concomitant miR-125b elevation. The data from these AD models highlight miR-21 as a promising biomarker and a disease-modifying target to be further explored.


Assuntos
Doença de Alzheimer , Exossomos , MicroRNAs , Animais , Humanos , Camundongos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Exossomos/genética , MicroRNAs/genética , Neuroblastoma , Neuroglia , Neurônios
2.
Front Pharmacol ; 13: 839790, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35422699

RESUMO

Small extracellular vesicles (sEVs) have ∼30-200 nm diameter size and may act as carriers of different cargoes, depending on the cell of origin or on the physiological/pathological condition. As endogenous nanovesicles, sEVs are important in intercellular communication and have many of the desirable features of an ideal drug delivery system. sEVs are naturally biocompatible, with superior targeting capability, safety profile, nanometric size, and can be loaded with both lipophilic and hydrophilic agents. Because of their biochemical and physical properties, sEVs are considered a promising strategy over other delivery vehicles in the central nervous system (CNS) since they freely cross the blood-brain barrier and they can be directed to specific nerve cells, potentiating a more precise targeting of their cargo. In addition, sEVs remain stable in the peripheral circulation, making them attractive nanocarrier systems to promote neuroregeneration. This review focuses on the recent progress in methods for manufacturing, isolating, and engineering sEVs that can be used as a therapeutic strategy to overcome neurodegeneration associated with pathologies of the CNS, with particular emphasis on Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis diseases, as well as on brain tumors.

3.
Cells ; 10(9)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34572073

RESUMO

Neuronal miRNA dysregulation may have a role in the pathophysiology of Alzheimer's disease (AD). miRNA(miR)-124 is largely abundant and a critical player in many neuronal functions. However, the lack of models reliably recapitulating AD pathophysiology hampers our understanding of miR-124's role in the disease. Using the classical human SH-SY5Y-APP695 Swedish neuroblastoma cells (SH-SWE) and the PSEN1 mutant iPSC-derived neurons (iNEU-PSEN), we observed a sustained upregulation of miR-124/miR-125b/miR-21, but only miR-124 was consistently shuttled into their exosomes. The miR-124 mimic reduced APP gene expression in both AD models. While miR-124 mimic in SH-SWE neurons led to neurite outgrowth, mitochondria activation and small Aß oligomer reduction, in iNEU-PSEN cells it diminished Tau phosphorylation, whereas miR-124 inhibitor decreased dendritic spine density. In exosomes, cellular transfection with the mimic predominantly downregulated miR-125b/miR-21/miR-146a/miR-155. The miR-124 inhibitor upregulated miR-146a in the two experimental cell models, while it led to distinct miRNA signatures in cells and exosomes. In sum, though miR-124 function may be dependent on the neuronal AD model, data indicate that keeping miR-124 level strictly controlled is crucial for proper neuronal function. Moreover, the iNEU-PSEN cellular model stands out as a useful tool for AD mechanistic studies and perhaps for the development of personalized therapeutic strategies.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/patologia , MicroRNAs/administração & dosagem , MicroRNAs/genética , Neuroblastoma/patologia , Neurônios/patologia , Presenilina-1/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Exossomos/genética , Exossomos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neurônios/metabolismo , Transdução de Sinais
4.
Glia ; 68(8): 1631-1642, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32463968

RESUMO

Microglia are CNS-resident cells involved in immune surveillance and maintenance of intercellular homeostasis, while also contributing to neurologic pathologies. MicroRNAs (miRNAs) are a class of small (~22 nucleotides) single-stranded noncoding RNAs that participate in gene regulation at the post-transcriptional level. miRNAs typically bind to the untranslated region (3' UTR) of RNAs. It has been shown that miRNAs are important players in controlling inflammation and that their abnormal expression is linked to cancer and ageing, and to the onset and progression of neurodegenerative disease. Furthermore, miRNAs participate in intercellular trafficking. Thus, miRNAs are released from cells in a free form, bound to proteins or packaged within extracellular vesicles (EVs), exerting paracrine and long distance signaling. In this review, recent findings on the role of miRNAs as drivers of microglia phenotypic changes and their cotribution in neurological disease are addressed. MAIN POINTS: miRNAs have a key role in microglia function/dysfunction, polarization, and restoration. Microglia are both a source and recipient of extracellular vesicles (EVs) containing miRNAs. Extracellular miRNAs may be found as soluble (free and EV cargo) and protein complexes.


Assuntos
Exossomos/metabolismo , MicroRNAs/metabolismo , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Animais , Vesículas Extracelulares/metabolismo , Humanos , Inflamação/metabolismo , Doenças Neurodegenerativas/genética
5.
Eur J Med Chem ; 172: 16-25, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-30939350

RESUMO

Herein we report novel hybrid compounds based on valproic acid and DNA-alkylating triazene moieties, 1, with therapeutic potential for glioblastoma multiforme chemotherapy. We identified hybrid compounds 1d and 1e to be remarkably more potent against glioma and more efficient in decreasing invasive cell properties than temozolomide and endowed with chemical and plasma stability. In contrast to temozolomide, which undergoes hydrolysis to release an alkylating metabolite, the valproate hybrids showed a low potential to alkylate DNA. Key physicochemical properties align for optimal CNS penetration, highlighting the potential of these effective triazene based-hybrids for enhanced anticancer chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Triazenos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Neoplasias Encefálicas/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Glioma/patologia , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Triazenos/síntese química , Triazenos/química , Células Tumorais Cultivadas
6.
Eur J Nutr ; 58(1): 113-130, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29151137

RESUMO

PURPOSE: Epidemiological and intervention studies have attempted to link the health effects of a diet rich in fruits and vegetables with the consumption of polyphenols and their impact in neurodegenerative diseases. Studies have shown that polyphenols can cross the intestinal barrier and reach concentrations in the bloodstream able to exert effects in vivo. However, the effective uptake of polyphenols into the brain is still regarded with some reservations. Here we describe a combination of approaches to examine the putative transport of blackberry-digested polyphenols (BDP) across the blood-brain barrier (BBB) and ultimate evaluation of their neuroprotective effects. METHODS: BDP was obtained by in vitro digestion of blackberry extract and BDP major aglycones (hBDP) were obtained by enzymatic hydrolysis. Chemical characterization and BBB transport of extracts were evaluated by LC-MSn. BBB transport and cytoprotection of both extracts was assessed in HBMEC monolayers. Neuroprotective potential of BDP was assessed in NT2-derived 3D co-cultures of neurons and astrocytes and in primary mouse cerebellar granule cells. BDP-modulated genes were evaluated by microarray analysis. RESULTS: Components from BDP and hBDP were shown to be transported across the BBB. Physiologically relevant concentrations of both extracts were cytoprotective at endothelial level and BDP was neuroprotective in primary neurons and in an advanced 3D cell model. The major canonical pathways involved in the neuroprotective effect of BDP were unveiled, including mTOR signaling and the unfolded protein response pathway. Genes such as ASNS and ATF5 emerged as novel BDP-modulated targets. CONCLUSIONS: BBB transport of BDP and hBDP components reinforces the health benefits of a diet rich in polyphenols in neurodegenerative disorders. Our results suggest some novel pathways and genes that may be involved in the neuroprotective mechanism of the BDP polyphenol components.


Assuntos
Barreira Hematoencefálica/metabolismo , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Rubus/metabolismo , Animais , Células Cultivadas , Cromatografia Líquida , Humanos , Técnicas In Vitro , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Fármacos Neuroprotetores/metabolismo , Extratos Vegetais/metabolismo , Reação em Cadeia da Polimerase , Polifenóis/metabolismo
7.
Biochimie ; 155: 67-82, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29857185

RESUMO

Exosome-mediated intercellular communication has been increasingly recognized as having a broad impact on Alzheimer's disease (AD) pathogenesis. Still, limited information exists regarding their "modus operandi", as it critically depends on exosomal cargo, environmental context and target cells. Therefore, a more thorough understanding of the role of exosomes from different cell types as mediators of neuroinflammation in AD context is a decisive step to open avenues for innovative and efficient therapies. In this study, we demonstrate that SH-SY5Y cells transfected with the Swedish mutant of APP695 (SHSwe) remarkably express increased inflammatory markers, combined with higher APP and Aß1-40 production, when compared to naïve SH-SY5Y (SH) cells. Although exerting an early clearance effect on extracellular APP and Aß accumulation when in co-culture with SHSwe cells, human CHME3 microglia gradually lose such property, and express both pro-inflammatory (iNOS, IL-1ß, TNF-α, MHC class II, IL-6) and pro-resolving genes (IL-10 and Arginase 1), while also evidence increased senescence-associated ß-galactosidase activity. Interestingly, upregulation of inflammatory-associated miRNA (miR)-155, miR-146a and miR-124 by SHSwe secretome shows to be time-dependent and to inversely correlate with their respective targets (SOCS-1, IRAK1 and C/EBP-α). We report that microglia also internalize exosomes released from SHSwe cells, which are enriched in miR-155, miR-146a, miR-124, miR-21 and miR-125b and recapitulate the cells of origin. Furthermore, we show that SHSwe-derived exosomes are capable of inducing acute and delayed microglial upregulation of TNF-α, HMGB1 and S100B pro-inflammatory markers, from which only S100B is found on their derived exosomes. Most importantly, our data reveal that miR-21 is a consistent biomarker that is found not only in SHSwe cells and in their released exosomes, but also in the recipient CHME3 microglia and derived exosomes. This work contributes to the increased understanding of neuron-microglia communication and exosome-mediated neuroinflammation in AD, while highlights miR-21 as a promising biomarker/target for therapeutic intervention.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Comunicação Celular , Citocinas/metabolismo , Exossomos/metabolismo , MicroRNAs/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Linhagem Celular Tumoral , Citocinas/genética , Exossomos/genética , Exossomos/patologia , Humanos , MicroRNAs/genética , Microglia/patologia , Neurônios/patologia
8.
Mol Neurobiol ; 55(5): 4207-4224, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28612258

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of unknown cause. Absence of specific targets and biomarkers compromise the development of new therapeutic strategies and of innovative tools to stratify patients and assess their responses to treatment. Here, we investigate changes in neuroprotective-neuroinflammatory actions in the spinal cord of SOD1 G93A mice, at presymptomatic and symptomatic stages to identify stage-specific biomarkers and potential targets. Results showed that in the presymptomatic stage, there are alterations in both astrocytes and microglia, which comprise decreased expression of GFAP and S100B and upregulation of GLT-1, as well as reduced expression of CD11b, M2-phenotype markers, and a set of inflammatory mediators. Reduced levels of Connexin-43, Pannexin-1, CCL21, and CX3CL1 further indicate the existence of a compromised intercellular communication. In contrast, in the symptomatic stage, increased markers of inflammation became evident, such as NF-κB/Nlrp3-inflammasome, Iba1, pro-inflammatory cytokines, and M1-polarizion markers, together with a decreased expression of M2-phenotypic markers. We also observed upregulation of the CX3CL1-CX3CR1 axis, Connexin-43, Pannexin-1, and of microRNAs (miR)-124, miR-125b, miR-146a and miR-21. Reduced motor neuron number and presence of reactive astrocytes with decreased GFAP, GLT-1, and GLAST further characterized this inflammatory stage. Interestingly, upregulation of miR-155 and downregulation of MFG-E8 appear as consistent biomarkers of both presymptomatic and symptomatic stages. We hypothesize that downregulated cellular interplay at the early stages may represent neuroprotective mechanisms against inflammation, SOD1 aggregation, and ALS onset. The present study identified a set of inflamma-miRNAs, NLRP3-inflammasome, HMGB1, CX3CL1-CX3CR1, Connexin-43, and Pannexin-1 as emerging candidates and promising pharmacological targets that may represent potential neuroprotective strategies in ALS therapy.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Regulação para Baixo , MicroRNAs/metabolismo , Neuroglia/patologia , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Comunicação Celular , Contagem de Células , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Proteína HMGB1/metabolismo , Homeostase , Humanos , Inflamassomos/metabolismo , Inflamação/patologia , Interleucina-6/metabolismo , Camundongos Transgênicos , MicroRNAs/genética , Neurônios Motores/metabolismo , Neurônios Motores/patologia , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neuroglia/metabolismo , Fenótipo , Transdução de Sinais , Superóxido Dismutase-1/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/genética
9.
ACS Chem Neurosci ; 8(1): 50-59, 2017 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-27665765

RESUMO

Glioblastoma (GBM) is the most common and aggressive type of brain tumor in adults. The triazene Temozolomide (TMZ), an alkylating drug, is the classical chemotherapeutic agent for gliomas, but has been disappointing against the highly invasive and resistant nature of GBM. Hybrid compounds may open new horizons within this challenge. The multicomponent therapeutic strategy here used resides on a combination of two repurposing drugs acting by different but potentially synergistic mechanisms, improved efficacy, and lower resistance effects. We synthesized a new hybrid compound (HYBCOM) by covalently binding a TMZ analogue to valproic acid, a histone deacetylase inhibitor drug that was shown to sensitize TMZ-resistant glioma cells. Advantages of this new molecule as compared to TMZ, in terms of chemotherapeutic efficacy, were investigated. Our results evidenced that HYBCOM more efficiently decreased the viability and proliferation of the GL261 glioma cells, while showing to better target the tumor cells than the functionally normal astrocytes. Increased cytotoxicity by HYBCOM may be a consequence of the improved autophagic process observed. Additionally, HYBCOM changed the morphology of GL261 cells into a nonpolar, more rounded shape, impairing cell migration ability. Most interesting, and in opposite to TMZ, cells exposed to HYBCOM did not enhance the expression of drug resistance proteins, a major issue in the treatment of GBM. Overall, our studies indicate that HYBCOM has promising chemotherapeutic benefits over the classical TMZ, and future studies should assess if the treatment translates into efficacy in glioblastoma experimental models and reveal clinical benefits in GBM patients.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos Alquilantes/síntese química , Antineoplásicos Alquilantes/química , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/química , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Glioma/patologia , Humanos , Temozolomida
10.
Food Chem ; 215: 274-83, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27542476

RESUMO

Neuroinflammation is an integral part of the neurodegeneration process inherent to several aging dysfunctions. Within the central nervous system, microglia are the effective immune cells, responsible for neuroinflammatory responses. In this study, raspberries were subjected to in vitro digestion simulation to obtain the components that result from the gastrointestinal (GI) conditions, which would be bioaccessible and available for blood uptake. Both the original raspberry extract and the gastrointestinal bioaccessible (GIB) fraction protected neuronal and microglia cells against H2O2-induced oxidative stress and lipopolysaccharide (LPS)-induced inflammation, at low concentrations. Furthermore, this neuroprotective capacity was independent of intracellular ROS scavenging mechanisms. We show for the first time that raspberry metabolites present in the GIB fraction significantly inhibited microglial pro-inflammatory activation by LPS, through the inhibition of Iba1 expression, TNF-α release and NO production. Altogether, this study reveals that raspberry polyphenols may present a dietary route to the retardation or amelioration of neurodegenerative-related dysfunctions.


Assuntos
Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Rubus , Peróxido de Hidrogênio/farmacologia , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Microglia/metabolismo , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Fenol/metabolismo , Polifenóis/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
11.
Brain Res ; 1651: 17-26, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27639810

RESUMO

Natural food sources constitute a promising source of new compounds with neuroprotective properties, once they have the ability to reach the brain. Our aim was to evaluate the brain accessibility of quercetin, epigallocatechin gallate (EGCG) and cyanidin-3-glucoside (C3G) in relation to their neuroprotective capability. Primary cortical neuron cultures were exposed to oxidative insult in the absence and presence of the selected compounds, and neuroprotection was assessed through evaluation of apoptotic-like and necrotic-like cell death. The brain accessibility of selected compounds was assessed using an optimised human blood-brain barrier model. The blood-brain barrier model was crossed rapidly by EGCG and more slowly by C3G, but not by quercetin. EGCG protected against oxidation-induced neuronal necrotic-like cell death by ~40%, and apoptosis by ~30%. Both quercetin and C3G were less effective, since only the lowest quercetin concentration was protective, and C3G only prevented necrosis by ~37%. Quercetin, EGCG and C3G effectively inhibited α-synuclein fibrillation over the relevant timescale applied here. Overall, EGCG seems to be the most promising neuroprotective compound. Thus, inclusion of this polyphenol in the diet might provide an affordable means to reduce the impact of neurodegenerative diseases.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Flavonoides/farmacologia , Flavonoides/farmacocinética , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/farmacocinética , Animais , Antocianinas/farmacocinética , Antocianinas/farmacologia , Antioxidantes/farmacocinética , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Catequina/análogos & derivados , Catequina/farmacocinética , Catequina/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Estabilidade de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Glucosídeos/farmacocinética , Glucosídeos/farmacologia , Humanos , Necrose/tratamento farmacológico , Necrose/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Multimerização Proteica/efeitos dos fármacos , Quercetina/farmacocinética , Quercetina/farmacologia , Ratos Wistar , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/metabolismo
12.
Mediators Inflamm ; 2016: 6986175, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28096568

RESUMO

Identification of mediators triggering microglia activation and transference of noncoding microRNA (miRNA) into exosomes are critical to dissect the mechanisms underlying neurodegeneration. We used lipopolysaccharide- (LPS-) induced N9 microglia activation to explore new biomarkers/signaling pathways and to identify inflammatory miRNA (inflamma-miR) in cells and their derived exosomes. Upregulation of iNOS and MHC-II (M1-markers) and downregulation of arginase 1, FIZZ1 (M2-markers), and CX3CR1 (M0/M2 polarization) confirmed the switch of N9 LPS-treated cells into the M1 phenotype, as described for macrophages/microglia. Cells showed increased proliferation, activated TLR4/TLR2/NF-κB pathway, and enhanced phagocytosis, further corroborated by upregulated MFG-E8. We found NLRP3-inflammasome activation in these cells, probably accounting for the increased extracellular content of the cytokine HMGB1 and of the MMP-9 we have observed. We demonstrate for the first time that the inflamma-miR profiling (upregulated miR-155 and miR-146a plus downregulated miR-124) in M1 polarized N9 cells, noticed by others in activated macrophages/microglia, was replicated in their derived exosomes, likely regulating the inflammatory response of recipient cells and dissemination processes. Data show that LPS-treated N9 cells behave like M1 polarized microglia/macrophages, while providing new targets for drug discovery. In particular, the study yields novel insights into the exosomal circulating miRNA during neuroinflammation important for emerging therapeutic approaches targeting microglia activation.


Assuntos
Arginase/metabolismo , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/citologia , Microglia/citologia , Receptores de Interleucina-8A/metabolismo , Animais , Biomarcadores/metabolismo , Morte Celular , Proliferação de Células , Citocinas/metabolismo , Exossomos/metabolismo , Proteína HMGB1/metabolismo , Inflamassomos/metabolismo , Lipopolissacarídeos , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , MicroRNAs/metabolismo , Microglia/metabolismo , Fagocitose , Fenótipo
13.
Mol Neurobiol ; 53(6): 3976-3991, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26184632

RESUMO

Multiple sclerosis (MS) pathology is characterized by neuroinflammation and demyelination. Recently, the inflammatory molecule S100B was identified in cerebrospinal fluid (CSF) and serum of MS patients. Although seen as an astrogliosis marker, lower/physiological levels of S100B are involved in oligodendrocyte differentiation/maturation. Nevertheless, increased S100B levels released upon injury may induce glial reactivity and oligodendrocyte demise, exacerbating tissue damage during an MS episode or delaying the following remyelination. Here, we aimed to unravel the functional role of S100B in the pathogenesis of MS. Elevated S100B levels were detected in the CSF of relapsing-remitting MS patients at diagnosis. Active demyelinating MS lesions showed increased expression of S100B and its receptor, the receptor for advanced glycation end products (RAGE), in the lesion area, while chronic active lesions displayed increased S100B in demyelinated areas with lower expression of RAGE in the rim. Interestingly, reactive astrocytes were identified as the predominant cellular source of S100B, whereas RAGE was expressed by activated microglia/macrophages. Using an ex vivo demyelinating model, cerebral organotypic slice cultures treated with lysophosphatidylcholine (LPC), we observed a marked elevation of S100B upon demyelination, which co-localized mostly with astrocytes. Inhibition of S100B action using a directed antibody reduced LPC-induced demyelination, prevented astrocyte reactivity and abrogated the expression of inflammatory and inflammasome-related molecules. Overall, high S100B expression in MS patient samples suggests its usefulness as a diagnostic biomarker for MS, while the beneficial outcome of its inhibition in our demyelinating model indicates S100B as an emerging therapeutic target in MS.


Assuntos
Terapia de Alvo Molecular , Esclerose Múltipla Recidivante-Remitente/sangue , Esclerose Múltipla Recidivante-Remitente/líquido cefalorraquidiano , Subunidade beta da Proteína Ligante de Cálcio S100/sangue , Subunidade beta da Proteína Ligante de Cálcio S100/líquido cefalorraquidiano , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Neutralizantes/farmacologia , Antígenos de Neoplasias/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Citocinas/metabolismo , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamassomos/metabolismo , Lisofosfatidilcolinas/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Esclerose Múltipla Recidivante-Remitente/diagnóstico , Proteína 3 que Contém Domínio de Pirina da Família NLR , Testes de Neutralização , Substância Branca/metabolismo , Substância Branca/patologia , Adulto Jovem
14.
Brain Struct Funct ; 221(5): 2777-99, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26068054

RESUMO

Axon growth and dendrite development are key processes for the establishment of a functional neuronal network. Adenosine, which is released by neurons and glia, is a known modulator of synaptic transmission but its influence over neuronal growth has been much less investigated. We now explored the action of adenosine A2A receptors (A2AR) upon neurite outgrowth, discriminating actions over the axon or dendrites, and the mechanisms involved. Morphometric analysis of primary cultures of cortical neurons from E18 Sprague-Dawley rats demonstrated that an A2AR agonist, CGS 21680, enhances axonal elongation and dendritic branching, being the former prevented by inhibitors of phosphoinositide 3-kinase, mitogen-activated protein kinase and phospholipase C, but not of protein kinase A. By testing the influence of a scavenger of BDNF (brain-derived neurotrophic factor) over the action of the A2AR agonist and the action of a selective A2AR antagonist over the action of BDNF, we could conclude that while the action of A2ARs upon dendritic branching is dependent on the presence of endogenous BDNF, the influence of A2ARs upon axonal elongation is independent of endogenous BDNF. In consonance with the action over axonal elongation, A2AR activation promoted a decrease in microtubule stability and an increase in microtubule growth speed in axonal growth cones. In conclusion, we disclose a facilitatory action of A2ARs upon axonal elongation and microtubule dynamics, providing new insights for A2ARs regulation of neuronal differentiation and axonal regeneration.


Assuntos
Axônios/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Dendritos/fisiologia , Neurônios/fisiologia , Receptor A2A de Adenosina/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , Axônios/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Microtúbulos/fisiologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fenetilaminas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo
15.
Basic Clin Pharmacol Toxicol ; 116(5): 398-413, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25287116

RESUMO

Rosmarinic acid is a polyphenolic compound and main constituent of Rosmarinus officinalis and has been shown to possess antioxidant and anti-inflammatory properties. We aimed to evaluate the anti-inflammatory properties of rosmarinic acid and of an extract of R. officinalis in local inflammation (carrageenin-induced paw oedema model in the rat), and further evaluate the protective effect of rosmarinic acid in rat models of systemic inflammation: liver ischaemia-reperfusion (I/R) and thermal injury models. In the local inflammation model, rosmarinic acid was administered at 10, 25 and 50 mg/kg (p.o.), and the extract was administered at 10 and 25 mg/kg (equivalent doses to rosmarinic acid groups) to male Wistar rats. Administration of rosmarinic acid and extract at the dose of 25 mg/kg reduced paw oedema at 6 hr by over 60%, exhibiting a dose-response effect, suggesting that rosmarinic was the main contributor to the anti-inflammatory effect. In the liver I/R model, rosmarinic acid was administered at 25 mg/kg (i.v.) 30 min. prior to the induction of ischaemia and led to the significant reduction in the serum concentration of transaminases (AST and ALT) and LDH. In the thermal injury model, rosmarinic acid was administered at 25 mg/kg (i.v.) 5 min. prior to the induction of injury and significantly reduced multi-organ dysfunction markers (liver, kidney, lung) by modulating NF-κB and metalloproteinase-9. For the first time, the anti-inflammatory potential of rosmarinic acid has been identified, as it causes a substantial reduction in inflammation, and we speculate that it might be useful in the pharmacological modulation of injuries associated to inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Cinamatos/farmacologia , Depsídeos/farmacologia , Edema/prevenção & controle , Inflamação/prevenção & controle , Extratos Vegetais/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Rosmarinus , Animais , Anti-Inflamatórios/isolamento & purificação , Antioxidantes/farmacologia , Biomarcadores/sangue , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Carragenina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Edema/sangue , Edema/induzido quimicamente , Edema/imunologia , Inflamação/sangue , Inflamação/etiologia , Inflamação/imunologia , Mediadores da Inflamação/sangue , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fitoterapia , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Ratos Wistar , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/imunologia , Explosão Respiratória/efeitos dos fármacos , Rosmarinus/química , Fatores de Tempo , Ácido Rosmarínico
16.
Shock ; 43(4): 369-78, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25394244

RESUMO

Glycogen synthase kinase 3 (GSK-3) is a serine-threonine kinase discovered decades ago to have an important role in glycogen metabolism. Today, we know that this kinase is involved in the regulation of many cell functions, including insulin signaling, specification of cell fate during embryonic development, and the control of cell division and apoptosis. Insulin and TDZD-8 (4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione) are inhibitors of GSK-3ß that have been shown to possess organ-protective effects in inflammatory-mediated organ injury models. We aimed to evaluate the cytoprotective effect of GSK-3ß inhibition on rat models of liver ischemia-reperfusion and thermal injury. In the liver ischemia-reperfusion model, TDZD-8 and insulin were administered at 5 mg/kg (i.v.) and 1.4 IU/kg (i.v.), respectively, 30 min before induction of ischemia and led to the significant reduction of the serum concentration of aspartate aminotransferase, alanine aminotransferase, γ-glutamyltransferase, and lactate dehydrogenase. Beneficial effects were found to be independent from blood glucose levels. In the thermal injury model, TDZD-8 was administered at 5 mg/kg (i.v.) 5 min before induction of injury and significantly reduced multiple organ dysfunction markers (liver, neuromuscular, and lung). In the lung, TDZD-8 reduced the histological signs of tissue injury, inflammatory markers (cytokines), and neutrophil chemotaxis/infiltration; reduced GSK-3ß, nuclear factor-κB, and Akt activation; reduced caspase-3 and metalloproteinase-9 activation. Our study provides a new insight on the beneficial effects of GSK-3ß inhibition on systemic inflammation and further elucidates the mechanism and pathway crosstalks by which TDZD-8 reduces the multiple organ injury elicited by thermal injury.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Quinase 3 da Glicogênio Sintase/metabolismo , Fígado/fisiopatologia , Traumatismo por Reperfusão , Animais , Aspartato Aminotransferases/metabolismo , Linhagem da Célula , Citocinas/sangue , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Temperatura Alta , Humanos , Inflamação/metabolismo , Insulina/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Neutrófilos/metabolismo , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Tiadiazóis/química
17.
J Neurosurg ; 121(1): 55-62, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24702327

RESUMO

UNLABELLED: OBJECT.: There is an unmet clinical need to develop neuroprotective agents for neurosurgical and endovascular procedures that require transient cerebral artery occlusion. The aim in this study was to explore the effects of a single dose of recombinant human erythropoietin (rhEPO) before middle cerebral artery (MCA) occlusion in a focal cerebral ischemia/reperfusion model. METHODS: Twenty-eight adult male Wistar rats were subjected to right MCA occlusion via the intraluminal thread technique for 60 minutes under continuous cortical perfusion monitoring by laser Doppler flowmetry. Rats were divided into 2 groups: control and treatment. In the treated group, rhEPO (1000 IU/kg intravenously) was administered 10 minutes before the onset of the MCA ischemia. At 24-hour reperfusion, animals were examined for neurological deficits, blood samples were collected, and animals were killed. The following parameters were evaluated: brain infarct volume, ipsilateral hemispheric edema, neuron-specific enolase plasma levels, parenchyma histological features (H & E staining), Fluoro-Jade-positive neurons, p-Akt and total Akt expression by Western blot analysis, and p-Akt-positive nuclei by immunohistochemical investigation. RESULTS: Infarct volume and Fluoro-Jade staining of degenerating neurons in the infarct area did not vary between groups. The severity of neurological deficit (p < 0.001), amount of brain edema (78% reduction in treatment group, p < 0.001), and neuron-specific enolase plasma levels (p < 0.001) were reduced in the treatment group. Perivascular edema was histologically less marked in the treatment group. No variations in the expression or localization of p-Akt were seen. CONCLUSIONS: Administration of rhEPO before the onset of 60-minute transient MCA ischemia protected the brain from this insult. It is unlikely that rhEPO pretreatment leads to direct neuronal antiapoptotic effects, as supported by the lack of Akt activation, and its benefits are most probably related to an indirect effect on brain edema as a consequence of blood-brain barrier preservation. Although research on EPO derivatives is increasing, rhEPO acts through distinct neuroprotective pathways and its clinical safety profile is well known. Clinically available rhEPO is a potential therapy for prevention of neuronal injury induced by transitory artery occlusion during neurovascular procedures.


Assuntos
Encéfalo/efeitos dos fármacos , Eritropoetina/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Eritropoetina/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Fármacos Neuroprotetores/farmacologia , Fosfopiruvato Hidratase/sangue , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar
18.
Neurotox Res ; 26(1): 1-15, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24122290

RESUMO

Previous studies using monotypic nerve cell cultures have shown that bilirubin-induced neurological dysfunction (BIND) involves apoptosis and necrosis-like cell death, following neuritic atrophy and astrocyte activation,and that glycoursodeoxycholic acid (GUDCA) has therapeutic efficacy against BIND. Cross-talk between neurons and astrocytes may protect or aggravate neurotoxicity by unconjugated bilirubin (UCB). In a previous work we have shown that bidirectional signaling during astrocyte-neuron recognition attenuates neuronal damage by UCB. Here, we investigated whether the establishment of neuron-astrocyte homeostasis prior to cell exposure to UCB was instead associated with a lower resistance of neurons to UCB toxicity, and if the pro-survival properties of GUDCA were replicated in that experimental model. We have introduced a 24 h adaptation period for neuron-glia communication prior to the 48 h treatment with UCB. In such conditions, UCB induced glial activation, which aggravated neuronal damage, comprising increased apoptosis,cell demise and neuritic atrophy, which were completely prevented in the presence of GUDCA. Neuronal multidrug resistance-associated protein 1 expression and tumor necrosis factor-a secretion, although unchanged by UCB, increased in the presence of astrocytes. The rise in S100B and nitric oxide in the co-cultures medium may have contributed to UCB neurotoxicity. Since the levels of these diffusible molecules did not change by GUDCA we may assume that they are not directly involved in its beneficial effects. Data indicate that astrocytes, in an indirect neuron-astrocyte co-culture model and after homeostatic setting regulation of the system, are critically influencing neurodegeneration by UCB, and support GUDCA for the prevention of BIND.


Assuntos
Antioxidantes/toxicidade , Astrócitos/efeitos dos fármacos , Bilirrubina/toxicidade , Comunicação Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Astrócitos/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Comunicação Celular/fisiologia , Técnicas de Cultura de Células , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/metabolismo , Ratos Wistar , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ácido Ursodesoxicólico/análogos & derivados , Ácido Ursodesoxicólico/farmacologia
19.
Mol Neurobiol ; 49(1): 424-39, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23982745

RESUMO

The cerebellum is one of the most affected brain regions in the course of bilirubin-induced neurological dysfunction. We recently demonstrated that unconjugated bilirubin (UCB) reduces oligodendrocyte progenitor cell (OPC) survival and impairs oligodendrocyte (OL) differentiation and myelination in co-cultures of dorsal root ganglia neurons and OL. Here, we used organotypic cerebellar slice cultures, which replicate many aspects of the in vivo system, to dissect myelination defects by UCB in the presence of neuroimmune-related glial cells. Our results demonstrate that treatment of cerebellar slices with UCB reduces the number of myelinated fibres and myelin basic protein mRNA expression. Interestingly, UCB addition to slices increased the percentage of OPC and decreased mature OL content, whereas it decreased Olig1 and increased Olig2 mRNA expression. These UCB effects were associated with enhanced gliosis, revealed by an increased burden of both microglia and astrocytes. Additionally, UCB treatment led to a marked increase of tumor necrosis factor (TNF)-α and glutamate release, in parallel with a decrease of interleukin (IL)-6. No changes were observed relatively to IL-1ß and S100B secretion. Curiously, both α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist and TNF-α antibody partially prevented the myelination defects that followed UCB exposure. These data point to a detrimental role of UCB in OL maturation and myelination together with astrocytosis, microgliosis, and both inflammatory and excitotoxic responses, which collectively may account for myelin deficits following moderate to severe neonatal jaundice.


Assuntos
Bilirrubina/toxicidade , Cerebelo/metabolismo , Gliose/metabolismo , Bainha de Mielina/metabolismo , Receptores de AMPA/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Gliose/induzido quimicamente , Bainha de Mielina/efeitos dos fármacos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/metabolismo , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Receptores de AMPA/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
20.
Pediatr Neurol ; 49(6): 431-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24138949

RESUMO

BACKGROUND: Kernicterus is an irreversible brain damage caused by bilirubin deposition in selective brain regions. Sick and preterm infants with hyperbilirubinemia are particularly susceptible to the condition. METHODS: We studied autopsied brain tissue from a premature female infant with kernicterus with a bilirubin:albumin molar ratio of 1.0, hypoxia, acidosis, and seizures. The patient, previously described as having cerebellar axon/myelin loss and angiogenic sprouting, was assessed for histopathological features in brain regions less investigated, such as hippocampus and corpus striatum. Results were compared with age-matched controls. RESULTS: Increased blood vessel density with poorly defined lumen structures was observed in the mesencephalon, pons, and medulla oblongata, and, more predominantly, in the corpus striatum and hippocampus. These two regions exhibited increased expression of vascular endothelial growth factor, paralleled by vascular endothelial growth factor receptor-2, and albumin extravasation into the brain parenchyma. No similar findings were observed in the nonjaundiced babies with hypoxia that served as controls (one preterm with sepsis and a term infant with pneumonia). We found increased cellular expression of multidrug resistance-associated protein 1 and P-glycoprotein in the hippocampus, known as defensive mechanisms against bilirubin-induced cytotoxicity. Increased density of blood vessels and microvascular permeability, together with parenchymal albumin, may have contributed to increasing the brain content and retention of bilirubin, a condition implicated in kernicterus disease. CONCLUSIONS: This novel finding in a premature baby with kernicterus and associated risk factors deserves to be investigated in similar patients to better understand the less-well described effects of bilirubin-induced neurological sequelae in preterm infants.


Assuntos
Encéfalo/patologia , Kernicterus/diagnóstico , Nascimento Prematuro , Albuminas/metabolismo , Antígenos CD34/metabolismo , Autopsia , Vasos Sanguíneos/patologia , Encéfalo/metabolismo , Claudina-5/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Lactente , Masculino , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA