Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Int J Mol Sci ; 25(5)2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38474295

RESUMEN

Astroglia constitute the largest group of glial cells and are involved in numerous actions that are critical to neuronal development and functioning, such as maintaining the blood-brain barrier, forming synapses, supporting neurons with nutrients and trophic factors, and protecting them from injury. These properties are deeply affected in the course of many neurodegenerative diseases, including tauopathies, often before the onset of the disease. In this respect, the transfer of essential amino acids such as glutamate and glutamine between neurons and astrocytes in the glutamate-glutamine cycle (GGC) is one example. In this review, we focus on the GGC and the disruption of this cycle in tau-dependent neurodegeneration. A profound understanding of the complex functions of the GGC and, in the broader context, searching for dysfunctions in communication pathways between astrocytes and neurons via GGC in health and disease, is of critical significance for the development of novel mechanism-based therapies for neurodegenerative disorders.


Asunto(s)
Astrocitos , Ácido Glutámico , Glutamina , Enfermedades Neurodegenerativas , Neuronas , Humanos , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Comunicación Celular
2.
Int J Mol Sci ; 24(20)2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37895066

RESUMEN

In recent years, efforts have been made to identify new anti-cancer therapies. Various types of nanomaterials, including silver nanoparticles (AgNPs), are being considered as an option. In addition to its well-known antibacterial activity, AgNPs exhibit cytotoxic potential in both physiological and cancer cells by inducing stress-mediated autophagy and apoptotic cell death. A rapidly growing collection of data suggests that the proper regulation of autophagic machinery may provide an efficient tool for suppressing the development of cancer. In this light, AgNPs have emerged as a potential anti-cancer agent to support therapy of the disease. This review summarizes current data indicating the dual role of AgNP-induced autophagy and highlights factors that may influence its protective vs. its toxic potential. It also stresses that our understanding of the cellular and molecular mechanisms of autophagy machinery in cancer cells, as well as AgNP-triggered autophagy in both normal and diseased cells, remains insufficient.


Asunto(s)
Nanopartículas del Metal , Nanopartículas del Metal/toxicidad , Plata/toxicidad , Autofagia , Autofagosomas/metabolismo , Línea Celular Tumoral , Apoptosis
3.
Int J Mol Sci ; 24(17)2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37685956

RESUMEN

Glutamine (Gln), glutamate (Glu), and γ-amino butyric acid (GABA) are essential amino acids for brain metabolism and function. Astrocyte-derived Gln is the precursor for the two most important neurotransmitters in the central nervous system (CNS), which are the excitatory neurotransmitter Glu and the inhibitory neurotransmitter GABA. In addition to their roles in neurotransmission, these amino acids can be used as alternative substrates in brain metabolism that enable metabolic coupling between astrocytes and neurons in the glutamate-glutamine cycle (GGC). The disturbed homeostasis of these amino acids within the tripartite synapse may be involved in the pathogenesis of various neurological diseases. Interactions between astrocytes and neurons in terms of Gln, Glu, and GABA homeostasis were studied in different phases of experimental allergic encephalomyelitis (EAE) in Lewis rats. The results of the study showed a decrease in the transport (uptake and release) of Gln and GABA in both neuronal and astrocyte-derived fractions. These effects were fully or partially reversed when the EAE rats were treated with memantine, a NMDA receptor antagonist. Changes in the expression and activity of selected glutamine/glutamate metabolizing enzymes, such as glutamine synthase (GS) and phosphate-activated glutaminase (PAG), which were affected by memantine, were observed in different phases of EAE. The results suggested perturbed homeostasis of Gln, Glu, and GABA during EAE, which may indicate alterations in neuron-astrocyte coupling and dysfunction of the tripartite synapse. Memantine appears to partially regulate the disturbed relationships between Gln, Glu, and GABA.


Asunto(s)
Antifibrinolíticos , Encefalomielitis Autoinmune Experimental , Animales , Ratas , Ratas Endogámicas Lew , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Glutamina , Memantina/farmacología , Memantina/uso terapéutico , Encéfalo , Ácido Glutámico , Ácido gamma-Aminobutírico , Aminoácidos , Homeostasis
4.
Int J Mol Sci ; 23(21)2022 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-36361797

RESUMEN

The growing production of silver nanoparticles (AgNPs), and their widespread use in medical and consumer products, poses a potential threat to the environment and raises questions about biosafety. Immature organisms are particularly susceptible to various insults during development. The biological characteristics of immature organisms are different from those of adults, and dictate the consequences of exposure to various toxic substances, including AgNPs. Nanoparticles are highly reactive and can easily cross the blood-brain barrier (BBB) to accumulate in brain tissues. It is therefore important to investigate the molecular mechanisms of AgNP-induced neurotoxicity in the developing brain. Immature 2-week-old rats were exposed to a low dose of AgNPs (0.2 mg/kg b.w.) over a long period. Subsequently, brain tissues of the animals were subjected to ultrastructural and molecular analyses to determine endoplasmic reticulum (ER) stress. Ultrastructural markers of ER stress, such as pathological alterations in the ER and elongated forms of mitochondria accompanied by autophagy structures, were confirmed to be present in AgNP-exposed rat brain. Evidence for induction of ER stress in neurons was also provided by molecular markers. Upregulation of genes related to the ER-stress-induced unfolded protein response (UPR) pathway, such as GRP78, PERK, and CHOP ATF-6, was observed at the transcriptional and translational levels. The results show that prolonged exposure of immature rats to a low dose of AgNPs during the developmental period leads to induction of ER stress in the neurons of the developing brain. Simultaneously, in response to AgNP-induced ER stress, neurons promote protective mechanisms that partially compensate for ER stress by regulating the biodynamic processes of mitochondria and autophagy.


Asunto(s)
Estrés del Retículo Endoplásmico , Nanopartículas del Metal , Animales , Ratas , Nanopartículas del Metal/toxicidad , Nanopartículas del Metal/química , Plata/química , Respuesta de Proteína Desplegada , Encéfalo/metabolismo , Apoptosis
5.
Eur J Neurosci ; 54(9): 7377-7404, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34561918

RESUMEN

Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system which leads to neurological dysfunctions and severe disabilities. MS pathology is characterised by damage of the blood-brain barrier and infiltration of autoreactive T cells that overactivate glial cells, thereby initiating neuroinflammation accompanied by the formation of demyelinating plaques and neurodegeneration. Clinical deficits in this multifactorial disease depend on the progression of myelin loss, the stage of inflammation, the status of axons and the activity of oligodendrocyte precursor cells (OPCs). Despite significant progress in the treatment of MS, current therapies remain limited and new approaches are highly desirable. Nanosystems based on liposomes and nanoparticles are among some of the more noteworthy therapeutic strategies being investigated. Applications of nanosystems alone or as drug carriers in animal models of MS have been found to successfully alleviate the symptoms of the disease and exert anti-inflammatory potential. Exosomes are a specific type of nanosystem based on nanometre-sized extracellular vesicles released by different cells which exhibit important healing features. Exosomes contain an array of anti-inflammatory and neuroprotective agents which may contribute to modulation of the immune system as well as promoting remyelination and tissue repair. In this review, opportunities to use nanosystems against progression of MS will be discussed in context of cell-specific pathologies associated with MS.


Asunto(s)
Exosomas , Esclerosis Múltiple , Remielinización , Animales , Sistema Nervioso Central , Esclerosis Múltiple/tratamiento farmacológico , Vaina de Mielina , Neuroglía , Oligodendroglía
6.
Ecotoxicol Environ Saf ; 208: 111416, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33075587

RESUMEN

The increasing production and use of silver nanoparticles (AgNPs) as antimicrobial agents in medicinal and commercial products creates a substantial risk of exposure, especially for infants and children. Our current knowledge concerning the impact of AgNPs on developing brain is insufficient. Therefore we investigated the temporal profile of transcriptional changes in cellular components of the neurovascular unit in immature rats exposed to a low dose of AgNPs. The behavior of animals under these conditions was also monitored. Significant deposition of AgNPs in brain of exposed rats was identified and found to persist over the post-exposure time. Substantial changes were noted in the transcriptional profile of tight junction proteins such as occludin and claudin-5, and pericyte-related molecules such as angiopoietin-1. Moreover, downregulation of platelet-derived growth factor (PDGFß) and its receptor (PDGFßR) which constitute the main signaling pathway between endothelial cells and pericytes was observed. These were long-lasting effects, accompanied by overexpression of astroglial-specific GFAP mRNA and endothelial cell adhesion molecule, ICAM-1, involved in the pathomechanism of neuroinflammation. The profile of changes indicates that even low doses of AgNPs administered during the early stage of life induce dysregulation of neurovascular unit constituents which may lead to disintegration of the blood-brain barrier. This was confirmed by ultrastructural analysis that revealed enhanced permeability of cerebral microvessels resulting in perivascular edema. Changes in the behavior of exposed rats indicating pro-depressive and anti-anxiety impacts were also identified. The results show a high risk of using AgNPs in medical and consumer products dedicated for infants and children.


Asunto(s)
Nanopartículas del Metal/toxicidad , Plata/toxicidad , Angiopoyetina 1 , Animales , Astrocitos/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , Claudina-5/genética , Claudina-5/metabolismo , Claudina-5/farmacología , Células Endoteliales/metabolismo , Masculino , Nanopartículas del Metal/química , Microvasos , Ocludina/genética , Permeabilidad , Ratas , Plata/química , Pruebas de Toxicidad , Transcriptoma
7.
Int J Mol Sci ; 22(16)2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34445109

RESUMEN

Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system that leads to the progressive disability of patients. A characteristic feature of the disease is the presence of focal demyelinating lesions accompanied by an inflammatory reaction. Interactions between autoreactive immune cells and glia cells are considered as a central mechanism underlying the pathology of MS. A glia-mediated inflammatory reaction followed by overproduction of free radicals and generation of glutamate-induced excitotoxicity promotes oligodendrocyte injury, contributing to demyelination and subsequent neurodegeneration. Activation of purinergic signaling, in particular P2X7 receptor-mediated signaling, in astrocytes and microglia is an important causative factor in these pathological processes. This review discusses the role of astroglial and microglial cells, and in particular glial P2X7 receptors, in inducing MS-related neuroinflammatory events, highlighting the importance of P2X7R-mediated molecular pathways in MS pathology and identifying these receptors as a potential therapeutic target.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animales , Humanos , Neuroglía/metabolismo , Transducción de Señal/fisiología
8.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34768760

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is an animal model most commonly used in research on the pathomechanisms of multiple sclerosis (MS). The inflammatory processes, glutamate excitotoxicity, and oxidative stress have been proposed as determinants accompanying demyelination and neuronal degeneration during the course of MS/EAE. The aim of the current study was to characterize the role of NMDA receptors in the induction of oxidative stress during the course of EAE. The effect of memantine, the uncompetitive NMDA receptor antagonist, on modulation of neurological deficits and oxidative stress in EAE rats was analyzed using several experimental approaches. We demonstrated that the expression of antioxidative enzymes (superoxide dismutases SOD1 and SOD2) were elevated in EAE rat brains. Under the same experimental conditions, we observed alterations in oxidative stress markers such as increased levels of malondialdehyde (MDA) and decreased levels of sulfhydryl (-SH) groups, both protein and non-protein (indicating protein damage), and a decline in reduced glutathione. Importantly, pharmacological inhibition of ionotropic NMDA glutamate receptors by their antagonist memantine improved the physical activity of EAE rats, alleviated neurological deficits such as paralysis of tail and hind limbs, and modulated oxidative stress parameters (MDA, -SH groups, SOD's). Furthermore, the current therapy aiming to suppress NMDAR-induced oxidative stress was partially effective when NMDAR's antagonist was administered at an early (asymptomatic) stage of EAE.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Memantina/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Femenino , Memantina/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Ratas Endogámicas Lew , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
9.
Int J Mol Sci ; 22(6)2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33802775

RESUMEN

Silver nanoparticles (AgNPs) are the one of the most extensively used nanomaterials. The strong antimicrobial properties of AgNPs have led to their use in a wide range of medical and consumer products. Although the neurotoxicity of AgNPs has been confirmed, the molecular mechanisms have not been extensively studied, particularly in immature organisms. Based on information gained from previous in vitro studies, in the present work, we examine whether ionotropic NMDA glutamate receptors contribute to AgNP-induced neurotoxicity in an animal model of exposure. In brains of immature rats subjected to a low dose of AgNPs, we identified ultrastructural and molecular alterations in the postsynaptic region of synapses where NMDA receptors are localized as a multiprotein complex. We revealed decreased expression of several NMDA receptor complex-related proteins, such as GluN1 and GluN2B subunits, scaffolding proteins PSD95 and SynGAP, as well as neuronal nitric oxide synthase (nNOS). Elucidating the changes in NMDA receptor-mediated molecular mechanisms induced by AgNPs, we also identified downregulation of the GluN2B-PSD95-nNOS-cGMP signaling pathway which maintains LTP/LTD processes underlying learning and memory formation during development. This observation is accompanied by decreased density of NMDA receptors, as assessed by a radioligand binding assay. The observed effects are reversible over the post-exposure time. This investigation reveals that NMDA receptors in immature rats are a target of AgNPs, thereby indicating the potential health hazard for children and infants resulting from the extensive use of products containing AgNPs.


Asunto(s)
Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Nanopartículas del Metal/toxicidad , Receptores de N-Metil-D-Aspartato/metabolismo , Plata/toxicidad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/ultraestructura , GMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Ligandos , Masculino , Nanopartículas del Metal/ultraestructura , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Subunidades de Proteína/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/ultraestructura
10.
Biochem J ; 476(22): 3493-3504, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31774919

RESUMEN

Astrocytes, by maintaining an optimal environment for neuronal function, play a critical role in proper function of mammalian nervous system. They regulate synaptic transmission and plasticity and protect neurons against toxic insults. Astrocytes and neurons interact actively via glutamine-glutamate cycle (GGC) that supports neuronal metabolic demands and neurotransmission. GGC deficiency may be involved in different diseases of the brain, where impaired astrocytic control of glutamate homeostasis contributes to neuronal dysfunction. This includes tau-dependent neurodegeneration, where astrocytes lose key molecules involved in regulation of glutamate/glutamine homeostasis, neuronal survival and synaptogenesis. Astrocytic dysfunction in tauopathy appears to precede neurodegeneration and overt tau neuropathology such as phosphorylation, aggregation and formation of neurofibrillary tangles. In this review, we summarize recent studies demonstrating that activation of astrocytes is strictly associated with neurodegenerative processes including those involved in tau related pathology. We propose that astrocytic dysfunction, by disrupting the proper neuron-glia signalling early in the disease, significantly contributes to tauopathy pathogenesis.


Asunto(s)
Astrocitos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas tau/metabolismo , Animales , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo
11.
Int J Mol Sci ; 21(23)2020 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-33256007

RESUMEN

Due to strong antimicrobial properties, silver nanoparticles (AgNPs) are used in a wide range of medical and consumer products, including those dedicated for infants and children. While AgNPs are known to exert neurotoxic effects, current knowledge concerning their impact on the developing brain is scarce. During investigations of mechanisms of neurotoxicity in immature rats, we studied the influence of AgNPs on glutamate transporter systems which are involved in regulation of extracellular concentration of glutamate, an excitotoxic amino acid, and compared it with positive control-Ag citrate. We identified significant deposition of AgNPs in brain tissue of exposed rats over the post-exposure time. Ultrastructural alterations in endoplasmic reticulum (ER) and Golgi complexes were observed in neurons of AgNP-exposed rats, which are characteristics of ER stress. These changes presumably underlie substantial long-lasting downregulation of neuronal glutamate transporter EAAC1, which was noted in AgNP-exposed rats. Conversely, the expression of astroglial glutamate transporters GLT-1 and GLAST was not affected by exposure to AgNPs, but the activity of the transporters was diminished. These results indicate that even low doses of AgNPs administered during an early stage of life create a substantial risk for health of immature organisms. Hence, the safety of AgNP-containing products for infants and children should be carefully considered.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Encéfalo/metabolismo , Nanopartículas del Metal/toxicidad , Plata/toxicidad , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/ultraestructura , Encéfalo/efectos de los fármacos , Transportador 3 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Plata/sangre , Sodio/metabolismo , Factores de Tiempo
12.
Inflammopharmacology ; 27(1): 129-137, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30209761

RESUMEN

Microglia-mediated neuroinflammation accompanies many central nervous system (CNS) diseases, including multiple sclerosis (MS), and is strongly dependent on the purinergic P2X7 receptor. The nature of the inflammatory response in MS is studied for decades indicating, that proinflammatory microgliosis is involved in advanced stages of MS and is associated with active tissue damage and neurological dysfunctions. Evidence on the role of microgliosis in initial stages of the disease is scarce. Thus, in the present study, we investigated the time course of microglial activation in rat brain subjected to experimental autoimmune encephalomyelitis (EAE) which is the animal model of MS. We show that activation of microglia occurs in brains of immunized rats at a very early stage of EAE, well before the development of neurological symptoms of the disease. Enhanced immunoreactivity of microglia/macrophage-specific protein Iba-1, together with morphological features of microgliosis, was identified beginning at day 4 post immunization. Concomitantly, microglial expression of P2X7R was also examined. Moreover, our results reveal that administration of Brilliant Blue G, an antagonist of P2X7R, delays the onset of the disease and partially inhibits development of neurological symptoms in EAE rats. Blockage of P2X7R significantly reduces activation of microglia as confirmed by decreased Iba-1 immunoreactivity and suppresses neuroinflammation in EAE rat brains, as indicated by decreased protein levels of investigated proinflammatory cytokines: IL-1ß, IL-6 and TNF-α. Our results indicate that microglia are involved in inducing neuroinflammation at a very early stage of MS/EAE via a P2X7R-dependent mechanism.


Asunto(s)
Encefalomielitis Autoinmune Experimental/fisiopatología , Microglía/metabolismo , Esclerosis Múltiple/fisiopatología , Receptores Purinérgicos P2X7/metabolismo , Animales , Encéfalo/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Cobayas , Esclerosis Múltiple/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas , Ratas Endogámicas Lew , Receptores Purinérgicos P2X7/efectos de los fármacos , Colorantes de Rosanilina/farmacología
13.
Purinergic Signal ; 14(4): 385-393, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30091000

RESUMEN

Purinergic P2X receptors, when activated under pathological conditions, participate in induction of the inflammatory response and/or cell death. Both neuroinflammation and neurodegeneration represent hallmarks of multiple sclerosis (MS), an autoimmune disease of the central nervous system. In the current study, we examined whether P2X7R is expressed in brain microvasculature of rats subjected to experimental autoimmune encephalomyelitis (EAE) and explore possible relationships with blood-brain barrier (BBB) protein-claudin-5 after administration of P2X7R antagonist-Brilliant Blue G (BBG). Capillary fraction isolated from control and EAE rat brains was subjected to immunohistochemical and Western blot analyses. We document the presence of P2X7R in brain capillaries isolated from brain tissue of EAE rats. P2X7R is found to be localized on the abluminal surface of the microvessels and is co-expressed with PDGFßR, a marker of pericytes. We also show over-expression of this receptor in isolated capillaries during the course of EAE, which is temporally correlated with a lower protein level of PDGFßR, as well as claudin-5, a tight junction-building protein. Administration of a P2X7R antagonist to the immunized rats significantly reduced clinical signs of EAE and enhances protein expression of both claudin-5 and PDGFßR. These results indicate that P2X7 receptor located on pericytes may contribute to pathological mechanisms operated during EAE in cerebral microvessels influencing the BBB integrity.


Asunto(s)
Capilares/efectos de los fármacos , Claudina-5/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/efectos de los fármacos , Animales , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Capilares/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Ratas , Receptores Purinérgicos P2X7/metabolismo
14.
Adv Exp Med Biol ; 1048: 227-250, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29453542

RESUMEN

The potent antimicrobial properties of nanoparticulate silver (AgNPs) have led to broad interest in using them in a wide range of commercial and medical applications. Although numerous in vivo and in vitro studies have provided evidence of toxic effects, rapid commercialization of AgNP-based nanomaterials has advanced without characterization of their potential environmental and health hazards. There is evidence that AgNPs can be translocated from the blood to the brain, regardless the route of exposure, and accumulate in the brain over time. As the brain is responsible for basic physiological functions and controls all human activities, it is important to assess the hazardous influence of AgNPs released from widely used nanoproducts and possible side effects of AgNP-based therapies. A number of studies have suggested that the size, shape and surface coating, as well as rates of silver ion release and interactions with proteins are the key factors determining the neurotoxicity of AgNPs. AgNPs target endothelial cells forming the blood-brain barrier, neurons and glial cells and leads finally to oxidative stress-related cell death. In this chapter, we review in detail current data on the impact of AgNPs on the central nervous system and discuss the possible mechanisms of their neurotoxic effects.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Nanopartículas del Metal/efectos adversos , Neuroglía , Neuronas , Estrés Oxidativo/efectos de los fármacos , Plata , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Muerte Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Plata/efectos adversos , Plata/farmacocinética
15.
Mol Cell Neurosci ; 74: 1-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26921791

RESUMEN

Astrocytes are the main cells responsible for maintenance of brain homeostasis. Undisturbed action and signaling with other cells are crucial for proper functioning of the central nervous system (CNS). Dysfunctional astrocytes may determine the degree of neuronal injury and are associated with several brain pathologies, among which are multiple sclerosis (MS) and the animal model of this disease which is known as experimental autoimmune encephalomyelitis (EAE). One of the many functions of astrocytes is their response to CNS damage when they undergo reactive gliosis. Our data reveal that activation of astrocytes occurs in forebrains of immunized rats at a very early stage of EAE, well before the symptomatic phase of the disease. We have noted enhanced expression of GFAP and S100ß starting from day 4 post-immunization. Temporal coincidence between the expression of astrocyte activation markers and the expression of connexin 43 and purinergic P2X7 receptor (P2X7R) was also observed. Administration of Brilliant blue G, an antagonist of P2X7R, significantly decreases astrogliosis as confirmed by immunohistochemical analysis and observation of decreased levels of GFAP and S100ß. The condition of the treated animals was improved and the neurological symptoms of the disease were alleviated. With the knowledge that cerebral astroglia represent the main source of ATP and glutamate which are potentially neurotoxic substances released through P2X7R and connexin hemichannels, we suggest that astroglia may be involved in pathogenesis of MS/EAE at a very early stage through the purinergic/glutamatergic mechanisms.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Gliosis/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animales , Astrocitos/efectos de los fármacos , Conexina 43/genética , Conexina 43/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Ratas , Ratas Endogámicas Lew , Colorantes de Rosanilina/farmacología , Colorantes de Rosanilina/uso terapéutico , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo
16.
Inflammopharmacology ; 25(4): 459-469, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28160128

RESUMEN

Kinins are bioactive peptides which provide multiple functions, including critical regulation of the inflammatory response. Released during tissue injury, kinins potentiate the inflammation which represents a hallmark of numerous neurological disorders, including those of autoimmune origin such as multiple sclerosis (MS). In the present work, we assess the expression of B1 receptor (B1R) in rat brain during the course of experimental autoimmune encephalomyelitis (EAE) which is an animal model of MS. We apply pharmacological inhibition to investigate the role of this receptor in the development of neurological deficits and in shaping the cytokine/chemokine profile during the course of the disease. Overexpression of B1R is observed in brain tissue of rats subjected to EAE, beginning at the very early asymptomatic phase of the disease. This overexpression is suppressed by a specific antagonist known as DALBK. The involvement of B1R in the progression of neurological symptoms in immunized rats is confirmed. Analysis of an array of cytokines/chemokines identified a sub-group as being B1R-dependent. Increase of the protein levels for the proinflammatory cytokines (Il-6, TNF-α but not IL-1ß), chemokines attracting immune cells into nervous tissue (MCP-1, MIP-3α, LIX), and protein levels of fractalkine and vascular endothelial growth factor observed in EAE rats, were significantly diminished after DALBK administration. This may indicate the protective potential of pharmacological inhibition of B1R. However, simultaneously reduced protein levels of anti-inflammatory and neuroprotective factors (IL-10, IL-4, and CNTF) was noticed. The results show that B1R-mediated signaling regulates the cellular response profile following neuroinflammation in EAE.


Asunto(s)
Antagonistas del Receptor de Bradiquinina B1/farmacología , Encéfalo/metabolismo , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/metabolismo , Receptor de Bradiquinina B1/biosíntesis , Animales , Bradiquinina/análogos & derivados , Bradiquinina/farmacología , Bradiquinina/uso terapéutico , Antagonistas del Receptor de Bradiquinina B1/uso terapéutico , Encéfalo/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Ratas , Ratas Endogámicas Lew
17.
Environ Toxicol ; 29(11): 1283-91, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23460582

RESUMEN

Silver nanoparticles, chemically neutral particles in the size range of 1-100 nm, express strong antimicrobial activity and therefore have a broad range of applications. The increased use of consumer products with nanosilver (nanoAg) may result in its release into the environment, and may particularly affect aquatic systems. The mechanisms of the harmful effects of nanoAg against aquatic organisms are unclear. Therefore, in the present study we investigate the pro-oxidative potential of these nanoparticles in experimentally exposed crayfish Orconectes limosus. Markers of oxidative stress and parameters of the antioxidant cell defense system such as total glutathione, glutathione reductase and the level of sulfhydryl groups were examined in the hepatopancreas of both sexes of O. limosus collected seasonally from Biale Lake (Poland) and subsequently exposed to nanoAg particles for 2 weeks. Exposure to nanoAg led to a high concentration-dependent increase in the rate of lipid peroxidation and a decrease of protein-bound SH groups which indicates protein oxidation. These markers of oxidative stress were accompanied by decreased levels of thiols and reduced activity of glutathione reductase. These results indicate a deficiency of reduced glutathione and suggest that the exposed organisms have less efficient antioxidative mechanisms available to counter ROS-mediated cellular stress. Furthermore, we find that confocal microscopy is of limited utility in monitoring the presence of silver nanoparticles in tissues of exposed crayfish.


Asunto(s)
Astacoidea/efectos de los fármacos , Hepatopáncreas/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Estrés Oxidativo/efectos de los fármacos , Plata/toxicidad , Animales , Antioxidantes/metabolismo , Astacoidea/metabolismo , Biomarcadores/metabolismo , Femenino , Glutatión/metabolismo , Glutatión Reductasa/metabolismo , Hepatopáncreas/metabolismo , Lagos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Polonia , Compuestos de Sulfhidrilo/metabolismo
18.
Cells ; 13(19)2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39404404

RESUMEN

Kinins are vasoactive peptides that are involved in various cellular mechanisms, including the inflammatory response. Kinins, released in vessel walls, exacerbate inflammation by modulating the production and release of pro-inflammatory factors via two types of G protein-related receptors-B1 and B2 receptors. B1 R is overexpressed during the inflammation that accompanies numerous neurological disorders, including multiple sclerosis (MS), in which loss of BBB integrity is an early pathomechanism of the disease. In this work, we apply pharmacological inhibition of the kinin B1 receptor with DALBK to investigate its effect on blood-brain barrier (BBB) permeability during the course of EAE, an animal model of MS. Functional, ultrastructural and molecular analyses were performed. The expression of selected BBB-associated proteins such as occludin and claudin-5 was assessed, as well as the astrocytic marker GFAP. We show that administration of a specific antagonist attenuates neurological symptoms in EAE rats and recovers the downregulation of TJ proteins and BBB leakage observed during the course of the disease, as well as significantly reducing the disease-specific activation of astroglia. The results show that B1 R-mediated signaling is involved in inducing molecular changes at the level of cerebral microvessels, leading to increased permeability of the BBB following neuroinflammation in EAE.


Asunto(s)
Barrera Hematoencefálica , Encefalomielitis Autoinmune Experimental , Microvasos , Receptor de Bradiquinina B1 , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Receptor de Bradiquinina B1/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Ratas , Microvasos/patología , Microvasos/metabolismo , Femenino , Permeabilidad Capilar , Bradiquinina/farmacología , Bradiquinina/metabolismo , Ocludina/metabolismo , Antagonistas del Receptor de Bradiquinina B1/farmacología , Astrocitos/metabolismo , Astrocitos/patología
19.
Folia Neuropathol ; 62(1): 1-12, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38741432

RESUMEN

Polychlorinated biphenyls (PCBs) and brominated flame retardants (BFRs) are dominant environmental and food contaminants. Tetrabromobisphenol A (TBBPA) is the most widely used BFR in the world to improve the fire safety of laminates in electrical and electronic equipment. Aroclor 1254, one of the PCBs, is widely distributed in the environment due to its extensive use in industrial applications around the world. Both groups of substances are potent toxicants. There is also increasing evidence that they have neurotoxic effects. In this study we tested the pro-inflammatory effects of Aroclor 1254 and TBBPA based on markers of microglial reactivity and levels of pro-inflammatory factors in the brain of immature rats. Aroclor 1254 or TBBPA were administered to the rats by oral gavage for two weeks at a dose of 10 mg/kg b.w. Both light and electron microscopy studies revealed features indicative of microglia activation in brains of exposed rats. Morphological changes were associated with overexpression of pro-inflammatory enzymes such as inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Analysis of cytokine/chemokine array revealed significant secretion of inflammatory mediators following exposure to both TBBPA and Aroclor 1254, which was stronger in the cerebellum than in the forebrain of exposed immature rats. The results indicate a pro-inflammatory profile of microglia activation as one of the neurotoxic mechanisms of both examined toxicants.


Asunto(s)
Microglía , Síndromes de Neurotoxicidad , Bifenilos Polibrominados , Animales , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Bifenilos Polibrominados/toxicidad , Ratas , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/etiología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/metabolismo , Masculino , Retardadores de Llama/toxicidad , Ratas Wistar
20.
Neurochem Int ; 168: 105551, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37295680

RESUMEN

Tau-dependent neurodegeneration is accompanied by astrocytosis in a mouse trans-genic model, which replicates the neuropathological characteristic of tauopathy and other human neurodegenerative disorders where astrocyte activation precedes neuronal loss and is associated with disease progression. This indicates an important role of astrocytes in the development of the disease. Astrocytes derived from a transgenic mouse model expressing human Tau, exhibit changes in cellular markers of astrocyte neuroprotective function related to the glutamate-glutamine cycle (GGC), representing a key part of astrocyte-neuron integrity. Here, we focused on investigating the functional properties of key GGC components involved in the astrocyte-neuron network associated with Tau pathology in vitro. Mutant recombinant Tau (rTau) carrying the P301L mutation was added to the neuronal cultures, with or without control astrocyte-conditioned medium (ACM), to study glutamine translocation through the GGC. We demonstrated that mutant Tau in vitro induces neuronal degeneration, while control astrocytes response in neuroprotective way by preventing neurodegeneration. In parallel with this observation, we noticed the Tau-dependent decline of neuronal microtubule associated protein 2 (MAP2), followed by changes in glutamine (Gln) transport. Exposure to rTau decreases sodium-dependent Gln uptake in neurons and that effect was reversed when cells were co-incubated with control ACM after induction of rTau dependent pathology. Further, we found that neuronal Na+-dependent system A is the most specific system that is affected under rTau exposure. In addition, in rTau-treated astrocytes total Na+-dependent uptake of Gln, which is mediated by the N system, increases. Altogether, our study suggest mechanisms operating in Tau pathology may be related to the alterations in glutamine transport and recycling that affect neuronal-astrocytic integrity.


Asunto(s)
Astrocitos , Tauopatías , Animales , Ratones , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Glutamina/metabolismo , Ratones Transgénicos , Neuronas/metabolismo , Proteínas tau/metabolismo , Tauopatías/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda