Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurochem Res ; 49(7): 1677-1686, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38451434

RESUMO

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease and the most common movement disorder. Although PD etiology is not fully understood, alpha (α)-synuclein is a key protein involved in PD pathology. MicroRNAs (miRNA), small gene regulatory RNAs that control gene expression, have been identified as biomarkers and potential therapeutic targets for brain diseases, including PD. In particular, miR-124 is downregulated in the plasma and brain samples of PD patients. Recently we showed that the brain delivery of miR-124 counteracts 6-hydroxydopamine-induced motor deficits. However, its role in α-synuclein pathology has never been addressed. Here we used paraquat (PQ)-induced rat PD model to evaluate the role of miR-124-3p in α-synuclein accumulation and dopaminergic neuroprotection. Our results showed that an intranigral administration of miR-124-3p reduced the expression and aggregation of α-synuclein in the substantia nigra (SN) of rats exposed to PQ. NADPH oxidases (NOX), responsible for reactive oxygen species generation, have been considered major players in the development of α-synuclein pathology. Accordingly, miR-124-3p decreased protein expression levels of NOX1 and its activator, small GTPase Rac1, in the SN of PQ-lesioned rats. Moreover, miR-124-3p was able to counteract the reduced levels of pituitary homeobox 3 (PITX3), a protein required for the dopaminergic phenotype, induced by PQ in the SN. This is the first study showing that miR-124-3p decreases PQ-induced α-synuclein levels and the associated NOX1/Rac1 signaling pathway, and impacts PITX3 protein levels, supporting the potential of miR-124-3p as a disease-modifying agent for PD and related α-synucleinopathies.


Assuntos
MicroRNAs , Paraquat , alfa-Sinucleína , Animais , MicroRNAs/metabolismo , alfa-Sinucleína/metabolismo , Paraquat/toxicidade , Masculino , Ratos , Ratos Wistar , Doença de Parkinson/metabolismo , Substância Negra/metabolismo , Substância Negra/efeitos dos fármacos , Modelos Animais de Doenças , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Ratos Sprague-Dawley
3.
J Neuroinflammation ; 19(1): 11, 2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-34991639

RESUMO

BACKGROUND: The brain vasculature plays a pivotal role in the inflammatory process by modulating the interaction between blood cells and the neurovascular unit. Argonaute-2 (Ago2) has been suggested as essential for endothelial survival but its role in the brain vasculature or in the endothelial-glial crosstalk has not been addressed. Thus, our aim was to clarify the significance of Ago2 in the inflammatory responses elicited by these cell types. METHODS: Mouse primary cultures of brain endothelial cells, astrocytes and microglia were used to evaluate cellular responses to the modulation of Ago2. Exposure of microglia to endothelial cell-conditioned media was used to assess the potential for in vivo studies. Adult mice were injected intraperitoneally with lipopolysaccharide (LPS) (2 mg/kg) followed by three daily intraperitoneal injections of Ago2 (0.4 nM) to assess markers of endothelial disruption, glial reactivity and neuronal function. RESULTS: Herein, we demonstrated that LPS activation disturbed the integrity of adherens junctions and downregulated Ago2 in primary brain endothelial cells. Exogenous treatment recovered intracellular Ago2 above control levels and recuperated vascular endothelial-cadherin expression, while downregulating LPS-induced nitric oxide release. Primary astrocytes did not show a significant change in Ago2 levels or response to the modulation of the Ago2 system, although endogenous Ago2 was shown to be critical in the maintenance of tumor necrosis factor-α basal levels. LPS-activated primary microglia overexpressed Ago2, and Ago2 silencing contained the inflammatory response to some extent, preventing interleukin-6 and nitric oxide release. Moreover, the secretome of Ago2-modulated brain endothelial cells had a protective effect over microglia. The intraperitoneal injection of LPS impaired blood-brain barrier and neuronal function, while triggering inflammation, and the subsequent systemic administration of Ago2 reduced or normalized endothelial, glial and neuronal markers of LPS damage. This outcome likely resulted from the direct action of Ago2 over the brain endothelium, which reestablished glial and neuronal function. CONCLUSIONS: Ago2 could be regarded as a putative therapeutic agent, or target, in the recuperation of the neurovascular unit in inflammatory conditions.


Assuntos
Proteínas Argonautas/farmacologia , Astrócitos/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Inativação Gênica , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/metabolismo
5.
Neurosci Lett ; 673: 116-121, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29518539

RESUMO

Perinatal stroke is often difficult to diagnose and an established treatment has not yet been validated, except for symptomatic measures. Herein, we propose to test the neuroprotective potential of the intravenous injection of retinoic acid-loaded nanoparticles (RA-NP) upon ischemic injury to the immature brain. The role of RA-NP on endothelial cells and organotypic slice cultures exposed to oxygen and glucose deprivation was assessed by evaluating markers pertaining to survival, proliferation, oxidative stress (NO, ROS), neuronal damage (enolase), vascular oxidation (p47phox) and microglia activation (CD68). Data showed that RA-NP (3 µg/ml) increased endothelial proliferation and survival, and normalized NO and ROS levels. The intravenous administration of RA-NP (10 µg/g) prevented ischemic injury in the hippocampus of 2-day-old mice by inhibiting cell death and normalizing markers of neurovascular function and inflammation. In sum, systemic administration of RA-NP protected neurovascular integrity and the inflammatory milieu from ischemia in the immature brain, highlighting its therapeutic value for perinatal stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/efeitos dos fármacos , Nanopartículas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Tretinoína/administração & dosagem , Administração Intravenosa , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Estresse Oxidativo
6.
J Tissue Eng Regen Med ; 12(1): e350-e354, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28182332

RESUMO

Stroke is a leading cause of death in adult life, closely behind ischemic heart disease, and causes a significant and abiding socioeconomic burden. However, current therapies are not able to ensure full neurologic and/or sequelae-free recovery to all stroke survivors. We believe treatment efficacy and patient rehabilitation could be enhanced significantly by targeting blood-brain barrier (BBB) deregulation and inflammation-induced barrier loss that occurs after stroke. In this pathological context, bone marrow-derived endothelial progenitor cells (EPC) enter the bloodstream towards the lesion site, but their insufficient numbers and impaired angiogenic ability compromise neurovascular regeneration. In this context, cell-based therapies have become increasingly appealing since treating patients with large numbers of mesenchymal or hematopoietic stem/progenitor cells alone may boost repair. However, this approach could be met with several challenges in terms of logistics and cost; hence, the development of a drug delivery system suitable for intravenous administration and functionalized for selective uptake by circulating EPC could enhance their restorative potential without perceived complications. The ability to encapsulate proangiogenic and anti-inflammatory agents, such as retinoic acid, and to safely and easily deliver them systemically may open new therapeutic perspectives for the treatment of cerebrovascular disorders. Copyright © 2017 John Wiley & Sons, Ltd.


Assuntos
Vasos Sanguíneos/fisiopatologia , Acidente Vascular Cerebral/terapia , Animais , Vasos Sanguíneos/patologia , Humanos , Nanomedicina , Regeneração
7.
Mediators Inflamm ; 2017: 6742427, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29138531

RESUMO

Inflammatory mechanisms triggered by microglial cells are involved in the pathophysiology of several brain disorders, hindering repair. Herein, we propose the use of retinoic acid-loaded polymeric nanoparticles (RA-NP) as a means to modulate microglia response towards an anti-inflammatory and neuroprotective phenotype (M2). RA-NP were first confirmed to be internalized by N9 microglial cells; nanoparticles did not affect cell survival at concentrations below 100 µg/mL. Then, immunocytochemical studies were performed to assess the expression of pro- and anti-inflammatory mediators. Our results show that RA-NP inhibited LPS-induced release of nitric oxide and the expression of inducible nitric oxide synthase and promoted arginase-1 and interleukin-4 production. Additionally, RA-NP induced a ramified microglia morphology (indicative of M2 state), promoting tissue viability, particularly neuronal survival, and restored the expression of postsynaptic protein-95 in organotypic hippocampal slice cultures exposed to an inflammatory challenge. RA-NP also proved to be more efficient than the free equivalent RA concentration. Altogether, our data indicate that RA-NP may be envisioned as a promising therapeutic agent for brain inflammatory diseases.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Microglia/efeitos dos fármacos , Nanopartículas/química , Tretinoína/química , Tretinoína/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
8.
J Neurochem ; 140(5): 692-702, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27925201

RESUMO

Angiogenesis and inflammation are clearly interconnected and interdependent processes that are dysregulated in a series of systemic and brain pathologies. Herein, key aspects regarding endothelial cell function and tissue remodelling that are particularly affected or aggravated by inflammation are presented. Most importantly, the cellular and molecular mechanisms involved in the vascular regulation of the inflammatory processes occurring in several brain disorders and how they impact on disease/injury progression are detailed, highlighting potential targets for therapy. Finally, nanomedicine-based approaches designed to overcome limitations pertaining to low systemic bioavailability, light, pH and temperature sensitivity and/or rapid degradation of these targets, and to optimize their mode of action are discussed. Ultimately, we expect this review to provide new insight and to suggest novel approaches for the treatment of blood-brain barrier dysfunction per se or as a means to treat the injured or diseased central nervous system.


Assuntos
Vasos Sanguíneos/fisiopatologia , Fármacos do Sistema Nervoso Central/uso terapêutico , Inflamação/fisiopatologia , Animais , Encefalopatias/tratamento farmacológico , Fármacos do Sistema Nervoso Central/farmacocinética , Endotélio Vascular/efeitos dos fármacos , Humanos , Neovascularização Fisiológica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...