Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37569690

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia worldwide, and it contributes up to 70% of cases. AD pathology involves abnormal amyloid beta (Aß) accumulation, and the link between the Aß1-42 structure and toxicity is of major interest. NMDA receptors (NMDAR) are thought to be essential in Aß-affected neurons, but the role of this receptor in glial impairment is still unclear. In addition, there is insufficient knowledge about the role of Aß species regarding mitochondrial redox states in neurons and glial cells, which may be critical in developing Aß-caused neurotoxicity. In this study, we investigated whether different Aß1-42 species-small oligomers, large oligomers, insoluble fibrils, and monomers-were capable of producing neurotoxic effects via microglial NMDAR activation and changes in mitochondrial redox states in primary rat brain cell cultures. Small Aß1-42 oligomers induced a concentration- and time-dependent increase in intracellular Ca2+ and necrotic microglial death. These changes were partially prevented by the NMDAR inhibitors MK801, memantine, and D-2-amino-5-phosphopentanoic acid (DAP5). Neither microglial intracellular Ca2+ nor viability was significantly affected by larger Aß1-42 species or monomers. In addition, the small Aß1-42 oligomers caused mitochondrial reactive oxygen species (mtROS)-mediated mitochondrial depolarization, glutamate release, and neuronal cell death. In microglia, the Aß1-42-induced mtROS overproduction was mediated by intracellular calcium ions and Aß-binding alcohol dehydrogenase (ABAD). The data suggest that the pharmacological targeting of microglial NMDAR and mtROS may be a promising strategy for AD therapy.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Ratas , Animales , Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Enfermedad de Alzheimer/metabolismo , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
2.
J Neurochem ; 154(3): 316-329, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31834946

RESUMEN

Tau is a microtubule-associated protein, found at high levels in neurons, and its aggregation is associated with neurodegeneration. Recently, it was found that tau can be actively secreted from neurons, but the effects of extracellular tau on neuronal viability are unclear. In this study, we investigated whether extracellular tau2N4R can cause neurotoxicity in primary cultures of rat brain neurons and glial cells. Cell cultures were examined for neuronal loss, death, and phosphatidylserine exposure, as well as for microglial phagocytosis by fluorescence microscopy. Aggregation of tau2N4R was assessed by atomic force microscopy. We found that extracellular addition of tau induced a gradual loss of neurons over 1-2 days, without neuronal necrosis or apoptosis, but accompanied by proliferation of microglia in the neuronal-glial co-cultures. Tau addition caused exposure of the 'eat-me' signal phosphatidylserine on the surface of living neurons, and this was prevented by elimination of the microglia or by inhibition of neutral sphingomyelinase. Tau also increased the phagocytic activity of pure microglia, and this was blocked by inhibitors of neutral sphingomyelinase or protein kinase C. The neuronal loss induced by tau was prevented by inhibitors of neutral sphingomyelinase, protein kinase C or the phagocytic receptor MerTK, or by eliminating microglia from the cultures. The data suggest that extracellular tau induces primary phagocytosis of stressed neurons by activated microglia, and identifies multiple ways in which the neuronal loss induced by tau can be prevented.


Asunto(s)
Microglía/efectos de los fármacos , Neuronas , Fagocitosis/efectos de los fármacos , Proteínas tau/farmacología , Animales , Células Cultivadas , Técnicas de Cocultivo , Microglía/metabolismo , Neuronas/patología , Ratas , Proteínas tau/metabolismo
3.
Molecules ; 25(9)2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397476

RESUMEN

The present study reports on the encapsulation of Elsholtzia ciliata ethanolic extract by freeze-drying method using skim milk, sodium caseinate, gum Arabic, maltodextrin, beta-maltodextrin, and resistant-maltodextrin alone or in mixtures of two or four encapsulants. The encapsulation ability of the final mixtures was evaluated based on their microencapsulating efficiency (EE) of total phenolic compounds (TPC) and the physicochemical properties of freeze-dried powders. Results showed that the freeze-dried powders produced using two encapsulants have a lower moisture content, but higher solubility, Carr index, and Hausner ratio than freeze-dried powders produced using only one encapsulant in the formulation. The microencapsulating efficiency of TPC also varied depending on encapsulants used. The lowest EE% of TPC was determined with maltodextrin (21.17%), and the highest with sodium caseinate (83.02%). Scanning electron microscopy revealed that freeze-drying resulted in the formation of different size, irregular shape glassy particles. This study demonstrated good mucoadhesive properties of freeze-dried powders, which could be incorporated in buccal or oral delivery dosage forms. In conclusion, the microencapsulation of E. ciliata ethanolic extract by freeze-drying is an effective method to produce new value-added pharmaceutical or food formulations with polyphenols.


Asunto(s)
Composición de Medicamentos/métodos , Lamiaceae/química , Fenoles/química , Animales , Cápsulas , Caseínas/química , Liofilización , Goma Arábiga/química , Microscopía Electrónica de Rastreo , Microtecnología , Leche/química , Tamaño de la Partícula , Extractos Vegetales/química , Polisacáridos/química
4.
Eur J Neurosci ; 50(2): 1994-2006, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30793394

RESUMEN

A growing number of studies suggest amyloid-ß and tau present in cerebrospinal fluid (CSF) and blood as putative biomarkers for Alzheimer's disease (AD). However, there is a question whether these compounds present in patients' bodily fluids can directly cause neurotoxic effects. We investigated effects of AD and other dementia (OD) patients' blood serum and CSF on viability of cells in primary cerebellar granule cell cultures. Overall, 59 individuals participated in the study from whom 55 samples of biological fluids were taken. Participants were classified into early (E-AD) and middle (M-AD) stages of AD, cognitively healthy control (HC) and OD groups. We found that concentrations of total and phosphorylated tau were higher in CSF from AD patients, while amyloid-ß42 and amyloid-ß40 in the serum was lower compared to HC. The most cytotoxic effects were induced by CSFs from M-AD patients which caused neuronal necrosis and suppressed microglial proliferation, whereas CSFs from the groups of other patients did not kill neurons. Serum and CSF from the E-AD group caused a reduction of neuronal numbers in cultures. There were no significant differences in levels of CSF biomarkers between the AD groups although both tau species in CSFs from M-AD patients were found to be significantly elevated compared to HC. Our data suggest that biological fluids from E-AD induce neuronal loss, whereas effects of CSF on the reduction in neuronal viability can serve as an indicator of M-AD and may be associated with extracellular tau.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Neuronas , Fragmentos de Péptidos/líquido cefalorraquídeo , Proteínas tau/líquido cefalorraquídeo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/sangre , Péptidos beta-Amiloides/sangre , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/sangre , Proteínas tau/sangre
5.
J Neurosci Res ; 93(3): 475-86, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25400096

RESUMEN

Although it is well documented that soluble beta amyloid (Aß) oligomers are critical factors in the pathogenesis of Alzheimer's disease (AD) by causing synaptic dysfunction and neuronal death, the primary mechanisms by which Aß oligomers trigger neurodegeneration are not entirely understood. We sought to investigate whether toxic small Aß(1-42) oligomers induce changes in plasma membrane potential of cultured neurons and glial cells in rat cerebellar granule cell cultures leading to neuronal death and whether these effects are sensitive to the N-methyl-D-aspartate receptor (NMDA-R) antagonist MK801. We found that small Aß(1-42) oligomers induced rapid, protracted membrane depolarization of both neurons and microglia, whereas there was no change in membrane potential of astrocytes. MK801 did not modulate Aß-induced neuronal depolarization. In contrast, Aß1(-42) oligomer-induced decrease in plasma membrane potential of microglia was prevented by MK801. Small Aß(1-42) oligomers significantly elevated extracellular glutamate and caused neuronal necrosis, and both were prevented by MK801. Also, small Aß(1-42) oligomers decreased resistance of isolated brain mitochondria to calcium-induced opening of mitochondrial permeability transition pore. In conclusion, the results suggest that the primary effect of toxic small Aß oligomers on neurons is rapid, NMDA-R-independent plasma membrane depolarization, which leads to neuronal death. Aß oligomers-induced depolarization of microglial cells is NMDA-R dependent.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Potenciales de la Membrana/efectos de los fármacos , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Muerte Celular/efectos de los fármacos , Cerebelo/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Microglía/metabolismo , Neuronas/metabolismo , Ratas
6.
J Neurochem ; 126(5): 604-15, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23745639

RESUMEN

Beta amyloid (Aß) oligomers are thought to contribute to the pathogenesis of Alzheimer's disease. However, clinical trials using Aß immunization were unsuccessful due to strong brain inflammation, the mechanisms of which are poorly understood. In this study we tested whether monoclonal antibodies to oligomeric Aß would prevent the neurotoxicity of Aß oligomers in primary neuronal-glial cultures. However, surprisingly,the antibodies dramatically increased the neurotoxicity of Aß. Antibodies bound to monomeric Aß fragments were non-toxic to cultured neurons, while antibodies to other oligomeric proteins: hamster polyomavirus major capsid protein, human metapneumovirus nucleocapsid protein, and measles virus nucleocapsid protein, strongly potentiated the neurotoxicity of their antigens. The neurotoxicity of antibody-antibody oligomeric antigen complexes was abolished by removal of the Fc region from the antibodies or by removal of microglia from cultures, and was accompanied by inflammatory activation and proliferation of the microglia in culture. In conclusion, we find that immune complexes formed by Aß oligomers or other oligomeric/multimeric antigens and their specific antibodies can cause death and loss of neurons in primary neuronal-glial cultures via Fc-dependent microglial activation. The results suggest that therapies resulting in antibodies to oligomeric Aß or oligomeric brain virus proteins should be used with caution or with suppression of microglial activation.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/toxicidad , Microglía/efectos de los fármacos , Síndromes de Neurotoxicidad/patología , Animales , Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
7.
J Biomed Sci ; 20: 10, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23432787

RESUMEN

BACKGROUND: The central molecule in the pathogenesis of Alzheimer's disease (AD) is believed to be a small-sized polypeptide - beta amyloid (Aß) which has an ability to assemble spontaneously into oligomers. Various studies concerning therapeutic and prophylactic approaches for AD are based on the immunotherapy using antibodies against Aß. It has been suggested that either active immunization with Aß or passive immunization with anti-Aß antibodies might help to prevent or reduce the symptoms of the disease. However, knowledge on the mechanisms of Aß-induced immune response is rather limited. Previous research on Aß1-42 oligomers in rat brain cultures showed that the neurotoxicity of these oligomers considerably depends on their size. In the current study, we evaluated the dependence of immunogenicity of Aß1-42 oligomers on the size of oligomeric particles and identified the immunodominant epitopes of the oligomers. RESULTS: Mice were immunized with various Aß1-42 oligomers. The analysis of serum antibodies revealed that small Aß1-42 oligomers (1-2 nm in size) are highly immunogenic. They induced predominantly IgG2b and IgG2a responses. In contrast, larger Aß1-42 oligomers and monomers induced weaker IgG response in immunized mice. The monoclonal antibody against 1-2 nm Aß1-42 oligomers was generated and used for antigenic characterization of Aß1-42 oligomers. Epitope mapping of both monoclonal and polyclonal antibodies demonstrated that the main immunodominant region of the 1-2 nm Aß1-42 oligomers is located at the amino-terminus (N-terminus) of the peptide, between amino acids 1 and 19. CONCLUSIONS: Small Aß1-42 oligomers of size 1-2 nm induce the strongest immune response in mice. The N-terminus of Aß1-42 oligomers represents an immunodominant region which indicates its surface localization and accessibility to the B cells. The results of the current study may be important for further development of Aß-based vaccination and immunotherapy strategies.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/administración & dosificación , Inmunidad Innata/efectos de los fármacos , Inmunoterapia , Fragmentos de Péptidos/administración & dosificación , Enfermedad de Alzheimer/tratamiento farmacológico , Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Antígenos/inmunología , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Mapeo Epitopo , Ratones , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Ratas
8.
Sci Rep ; 13(1): 10813, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37402829

RESUMEN

In tauopathies, abnormal deposition of intracellular tau protein followed by gradual elevation of tau in cerebrospinal fluids and neuronal loss has been documented, however, the mechanism how actually neurons die under tau pathology is largely unknown. We have previously shown that extracellular tau protein (2N4R isoform) can stimulate microglia to phagocytose live neurons, i.e. cause neuronal death by primary phagocytosis, also known as phagoptosis. Here we show that tau protein induced caspase-1 activation in microglial cells via 'Toll-like' 4 (TLR4) receptors and neutral sphingomyelinase. Tau-induced neuronal loss was blocked by caspase-1 inhibitors (Ac-YVAD-CHO and VX-765) as well as by TLR4 antibodies. Inhibition of caspase-1 by Ac-YVAD-CHO prevented tau-induced exposure of phosphatidylserine on the outer leaflet of neuronal membranes and reduced microglial phagocytic activity. We also show that suppression of NLRP3 inflammasome, which is down-stream of TLR4 receptors and mediates caspase-1 activation, by a specific inhibitor (MCC550) also prevented tau-induced neuronal loss. Moreover, NADPH oxidase is also involved in tau-induced neurotoxicity since neuronal loss was abolished by its pharmacological inhibitor. Overall, our data indicate that extracellular tau protein stimulates microglia to phagocytose live neurons via Toll-like 4 receptor-NLRP3 inflammasome-caspase-1 axis and NADPH oxidase, each of which may serve as a potential molecular target for pharmacological treatment of tauopathies.


Asunto(s)
Inflamasomas , Tauopatías , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas tau/metabolismo , Microglía/metabolismo , Caspasa 1/metabolismo , Receptor Toll-Like 4/metabolismo , Neuronas/metabolismo , Fagocitosis/fisiología , Tauopatías/metabolismo , NADPH Oxidasas/metabolismo
9.
Methods Mol Biol ; 2497: 63-72, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35771434

RESUMEN

Mitochondria participate in many physiological and pathological processes in the cells, including cellular energy supply, regulation of calcium homeostasis, apoptosis, and ROS generation. Alterations of mitochondrial functions, especially the opening of mitochondrial permeability transition pore (mPTP) are the main mechanisms responsible for the ischemic brain damage. Recently, the inhibitors of the Complex I of mitochondrial respiratory chain emerged as promising suppressors of mitochondrial ROS generation and mPTP opening. Here we describe the assay that can be implemented easily to evaluate the protective effects of rotenone or other potential inhibitors of the Complex I of mitochondrial respiratory chain against acute ischemia-induced injuries in brain.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial , Rotenona , Encéfalo/metabolismo , Calcio/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Humanos , Isquemia , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Necrosis por Permeabilidad de la Transmembrana Mitocondrial , Especies Reactivas de Oxígeno/metabolismo , Rotenona/farmacología
10.
Pharmaceutics ; 14(6)2022 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-35745831

RESUMEN

Essential oils contain many volatile compounds that are not stable and lose their pharmacological effect when exposed to the environment. The aim of this study is to protect Boswellia serrata L. essential oil from environmental factors by encapsulation and determine the influence of chitosan concentration and types (2%, 4%; medium and high molecular weights), essential oil concentration, different emulsifiers (Tween and Span), and technological factors (stirring time, launch height, drip rate) on the physical parameters, morphology, texture, and other parameters of the generated gels, emulsions, and microcapsules. For the first time, Boswellia serrata L. essential oil microcapsules with chitosan were prepared by coacervation. Hardness, consistency, stickiness, viscosity, and pH of chitosan gels were tested. Freshly obtained microcapsules were examined for moisture, hardness, resistance to compression, size, and morphology. Results show that different molecular weights and concentrations of chitosan affected gel hardness, consistency, stickiness, viscosity, mobility, and adhesion. An increase in chitosan concentration from 2% to 4% significantly changed the appearance of the microcapsules. It was found that spherical microcapsules were formed when using MMW and HMW 80/1000 chitosan. Chitosan molecular weight, concentration, essential oil concentration, and stirring time all had an impact on the hardness of the microcapsules and their resistance to compression.

11.
Biochim Biophys Acta ; 1797(6-7): 1012-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20381450

RESUMEN

We have previously reported that estradiol can protect heart mitochondria from the ischemia-induced mitochondrial permeability transition pore-related release of cytochrome c and subsequent apoptosis. In this study we investigated whether the effect of 17-beta-estradiol on ischemia-induced mitochondrial dysfunctions and apoptosis is mediated by activation of signaling protein kinases in a Langendorff-perfused rat heart model of stop-flow ischemia. We found that pre-perfusion of non-ischemic hearts with 100nM estradiol increased the resistance of subsequently isolated mitochondria to the calcium-induced opening of mitochondrial permeability transition pore and this was mediated by protein kinase G. Loading of the hearts with estradiol prevented ischemia-induced loss of cytochrome c from mitochondria and respiratory inhibition and these effects were reversed in the presence of the inhibitor of Akt kinase, NO synthase inhibitor L-NAME, guanylyl cyclase inhibitor ODQ and protein kinase G inhibitor KT5823. Estradiol prevented ischemia-induced activation of caspases and this was also reversed by KT5823. These findings suggest that estradiol may protect the heart against ischemia-induced injury activating the signaling cascade which involves Akt kinase, NO synthase, guanylyl cyclase and protein kinase G, and results in blockage of mitochondrial permeability transition pore-induced release of cytochrome c from mitochondria, respiratory inhibition and activation of caspases.


Asunto(s)
Caspasas/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Estradiol/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Carbazoles/farmacología , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Técnicas In Vitro , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Modelos Cardiovasculares , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
12.
Medicina (Kaunas) ; 47(2): 107-12, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21734444

RESUMEN

BACKGROUND AND OBJECTIVE. Alzheimer's disease is a progressive neurodegenerative disease that is biochemically characterized by the accumulation of amyloid beta (Aß) peptides in the brain. The current hypothesis suggests that Aß oligomers rather than fibrillar aggregates are the most toxic species of Aß though the mechanisms of their neurotoxicity are unclear. The authors have previously shown that small Aß(1-42) oligomers at around 1 µM concentration caused rapid (in 24 h) neuronal death in cerebellar granule cell (CGC) cultures. In this study, we aimed to investigate whether protracted (up to 7 days) incubation of CGC cultures with lower submicromolar concentration of various aggregates of Aß(1-42) had an effect on viability of neurons. In order to get some insight into the mechanism of Aß-induced cell death, we also sought to determine whether extracellular Ca(2+) and process of endocytosis contributed to Aß oligomer-induced neurotoxicity and whether pharmacological interventions into these processes would prevent Aß oligomer-induced cell death. MATERIAL AND METHODS. Primary cultures of CGC were treated with various aggregate forms of Aß(1-42). Cell viability was assessed by fluorescent microscopy using propidium iodide and Hoechst 33342 staining. RESULTS. Exposure of neurons to 500 nM Aß(1-42) oligomers for 72-168 h caused extensive neuronal necrosis. Lower concentrations (100-250 nM) were not toxic to cells during 7 days of incubation. Aß(1-42) monomers and fibrils had no effect on neuronal viability even after 7 days of incubation. Treatment of neurons with EGTA, steroid hormone 17ß-estradiol, and methyl-ß-cyclodextrin significantly reduced Aß(1-42) oligomers-induced neuronal death. CONCLUSIONS. The results show that submicromolar concentrations of Aß(1-42) oligomers were highly toxic to neurons during protracted incubation inducing neuronal necrosis that can be prevented by chelating extracellular Ca(2+) with EGTA, inhibiting endocytosis with methyl-ß-cyclodextrin, or by estradiol, which may protect against mitochondrial permeability transition pore opening.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Ácido Egtácico/farmacología , Endocitosis/efectos de los fármacos , Estradiol/farmacología , Neuronas/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Células Cultivadas , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Microscopía Fluorescente , Necrosis , Neuronas/metabolismo , Neuronas/patología , Propidio/farmacología , Ratas , Ratas Wistar , beta-Ciclodextrinas/farmacología
13.
Mol Neurobiol ; 58(2): 658-667, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33001416

RESUMEN

Recent data from various experimental models support the link between extracellular tau and neurodegeneration; however, the exact mechanisms by which extracellular tau or its modified forms or aggregates cause neuronal death remain unclear. We have previously shown that exogenously applied monomers and oligomers of the longest tau isoform (2N4R) at micromolar concentrations induced microglial phagocytosis of stressed-but-viable neurons in vitro. In this study, we investigated whether extracellular phosphorylated tau2N4R (p-tau2N4R), isoform 1N4R (tau1N4R) and K18 peptide can induce neuronal death or loss in primary neuronal-glial cell cultures. We found that p-tau2N4R at 30 nM concentration induced loss of viable neurons; however, 700 nM p-tau2N4R caused necrosis of both neurons and microglia, and this neuronal death was partially glial cell-dependent. We also found that extracellular tau1N4R oligomers, but not monomers, at 3 µM concentration caused neuronal death in mixed cell cultures: self-assembly tau1N4R dimers-tetramers induced neuronal necrosis and apoptosis, whereas Aß-promoted tau1N4R oligomers caused glial cell-dependent loss of neurons without signs of increased cell death. Monomeric and pre-aggregated tau peptide containing 4R repeats (K18) had no effect in mixed cultures, suggesting that tau neurotoxicity might be dependent on N-terminal part of the protein. Taken together, our results show that extracellular p-tau2N4R is the most toxic form among investigated tau species inducing loss of neurons at low nanomolar concentrations and that neurotoxicity of tau1N4R is dependent on its aggregation state.


Asunto(s)
Espacio Extracelular/química , Neuroglía/metabolismo , Neuronas/metabolismo , Neurotoxinas/toxicidad , Proteínas tau/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Masculino , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Agregado de Proteínas/efectos de los fármacos , Ratas Wistar
14.
Biology (Basel) ; 10(12)2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34943274

RESUMEN

Viral infections induce extracellular vesicles (EVs) containing viral material and inflammatory factors. Exosomes can easily cross the blood-brain barrier during respiratory tract infection and transmit the inflammatory signal to the brain; however, such a hypothesis has no experimental evidence. The study investigated whether exosome-like vesicles (ELVs) from virus mimetic poly (I:C)-primed airway cells enter the brain and interact with brain immune cells microglia. Airway cells were isolated from Wistar rats and BALB/c mice; microglial cell cultures-from Wistar rats. ELVs from poly (I:C)-stimulated airway cell culture medium were isolated by precipitation, visualised by transmission electron microscopy, and evaluated by nanoparticle analyser; exosomal markers CD81 and CD9 were determined by ELISA. For in vitro and in vivo tracking, particles were loaded with Alexa Fluor 555-labelled RNA. Intracellular reactive oxygen species (ROS) were evaluated by DCFDA fluorescence and mitochondrial superoxide-by MitoSOX. ELVs from poly (I:C)-primed airway cells entered the brain within an hour after intranasal introduction, were internalised by microglia and induced intracellular and intramitochondrial ROS production. There was no ROS increase in microglial cells was after treatment with ELVs from airway cells untreated with poly (I:C). In addition, poly (I:C)-primed airway cells induced inflammatory cytokine expression in the brain. The data indicate that ELVs secreted by virus-primed airway cells might enter the brain, cause the activation of microglial cells and neuroinflammation.

15.
Arch Biochem Biophys ; 496(2): 84-92, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20153288

RESUMEN

The link between the size of soluble amyloid beta (Abeta) oligomers and their toxicity to rat cerebellar granule cells (CGC) was investigated. Variation in conditions during in vitro oligomerization of Abeta(1-42) resulted in peptide assemblies with different particle size as measured by atomic force microscopy and confirmed by dynamic light scattering and fluorescence correlation spectroscopy. Small oligomers of Abeta(1-42) with a mean particle z-height of 1-2 nm exhibited propensity to bind to phospholipid vesicles and they were the most toxic species that induced rapid neuronal necrosis at submicromolar concentrations whereas the bigger aggregates (z-height above 4-5 nm) did not bind vesicles and did not cause detectable neuronal death. A similar neurotoxic pattern was also observed in primary cultures of cortex neurons whereas Abeta(1-42) oligomers, monomers and fibrils were non-toxic to glial cells in CGC cultures or macrophage J774 cells. However, both oligomeric forms of Abeta(1-42) induced reduction of neuronal cell densities in the CGC cultures.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Neuronas/efectos de los fármacos , Neuronas/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Peso Molecular , Ratas , Ratas Wistar , Relación Estructura-Actividad
16.
Biomolecules ; 10(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33019635

RESUMEN

Damage to cerebral mitochondria, particularly opening of mitochondrial permeability transition pore (MPTP), is a key mechanism of ischemic brain injury, therefore, modulation of MPTP may be a potential target for a neuroprotective strategy in ischemic brain pathologies. The aim of this study was to investigate whether biguanides-metformin and phenformin as well as other inhibitors of Complex I of the mitochondrial electron transfer system may protect against ischemia-induced cell death in brain slice cultures by suppressing MPTP, and whether the effects of these inhibitors depend on the age of animals. Experiments were performed on brain slice cultures prepared from 5-7-day (premature) and 2-3-month old (adult) rat brains. In premature brain slice cultures, simulated ischemia (hypoxia plus deoxyglucose) induced necrosis whereas in adult rat brain slice cultures necrosis was induced by hypoxia alone and was suppressed by deoxyglucose. Phenformin prevented necrosis induced by simulated ischemia in premature and hypoxia-induced-in adult brain slices, whereas metformin was protective in adult brain slices cultures. In premature brain slices, necrosis was also prevented by Complex I inhibitors rotenone and amobarbital and by MPTP inhibitor cyclosporine A. The latter two inhibitors were protective in adult brain slices as well. Short-term exposure of cultured neurons to phenformin, metformin and rotenone prevented ionomycin-induced MPTP opening in intact cells. The data suggest that, depending on the age, phenformin and metformin may protect the brain against ischemic damage possibly by suppressing MPTP via inhibition of mitochondrial Complex I.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Metformina/administración & dosificación , Mitocondrias/efectos de los fármacos , Fenformina/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Complejo I de Transporte de Electrón/genética , Humanos , Masculino , Mitocondrias/genética , Poro de Transición de la Permeabilidad Mitocondrial/antagonistas & inhibidores , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Necrosis por Permeabilidad de la Transmembrana Mitocondrial/efectos de los fármacos , Necrosis/tratamiento farmacológico , Necrosis/metabolismo , Necrosis/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fosforilación Oxidativa/efectos de los fármacos , Ratas
17.
Cell Death Dis ; 11(8): 645, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811809

RESUMEN

The pathogenesis of Alzheimer's disease (AD), a slowly-developing age-related neurodegenerative disorder, is a result of the action of multiple factors including deregulation of Ca2+ homeostasis, mitochondrial dysfunction, and dysproteostasis. Interaction of these factors in astrocytes, principal homeostatic cells in the central nervous system, is still poorly understood. Here we report that in immortalized hippocampal astrocytes from 3xTg-AD mice (3Tg-iAstro cells) bioenergetics is impaired, including reduced glycolysis and mitochondrial oxygen consumption, and increased production of reactive oxygen species. Shotgun proteomics analysis of mitochondria-ER-enriched fraction showed no alterations in the expression of mitochondrial and OxPhos proteins, while those related to the ER functions and protein synthesis were deregulated. Using ER- and mitochondria-targeted aequorin-based Ca2+ probe we show that, in 3Tg-iAstro cells, ER was overloaded with Ca2+ while Ca2+ uptake by mitochondria upon ATP stimulation was reduced. This was accompanied by the increase in short distance (≈8-10 nm) contact area between mitochondria and ER, upregulation of ER-stress/unfolded protein response genes Atf4, Atf6 and Herp, and reduction of global protein synthesis rate. We suggest that familial AD mutations in 3Tg-iAstro cells induce mitochondria-ER interaction changes that deregulate astrocytic bioenergetics, Ca2+ homeostasis and proteostasis. These factors may interact, creating a pathogenic loop compromising homeostatic and defensive functions of astroglial cells predisposing neurons to dysfunction.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Astrocitos/metabolismo , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Glucólisis/fisiología , Hipocampo/metabolismo , Homeostasis , Humanos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Neuronas/metabolismo , Consumo de Oxígeno/fisiología , Proteómica , Proteostasis , Especies Reactivas de Oxígeno/metabolismo , Respuesta de Proteína Desplegada
18.
J Biomed Sci ; 16: 70, 2009 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-19671187

RESUMEN

BACKGROUND: Heart ischemia can rapidly induce apoptosis and mitochondrial dysfunction via mitochondrial permeability transition-induced cytochrome c release. We tested whether nitric oxide (NO) can block this damage in isolated rat heart, and, if so, by what mechanisms. METHODS: Hearts were perfused with 50 microM DETA/NO (NO donor), then subjected to 30 min stop-flow ischemia or ischemia/reperfusion. Isolated heart mitochondria were used to measure the rate of mitochondrial oxygen consumption and membrane potential using oxygen and tetraphenylphosphonium-selective electrodes. Mitochondrial and cytosolic cytochrome c levels were measured spectrophotometrically and by ELISA. The calcium retention capacity of isolated mitochondria was measured using the fluorescent dye Calcium Green-5N. Apoptosis and necrosis were evaluated by measuring the activity of caspase-3 in cytosolic extracts and the activity of lactate dehydrogenase in perfusate, respectively. RESULTS: 30 min ischemia caused release of mitochondrial cytochrome c to the cytoplasm, inhibition of the mitochondrial respiratory chain, and stimulation of mitochondrial proton permeability. 3 min perfusion with 50 microM DETA/NO of hearts prior to ischemia decreased this mitochondrial damage. The DETA/NO-induced blockage of mitochondrial cytochrome c release was reversed by a protein kinase G (PKG) inhibitor KT5823, or soluble guanylate cyclase inhibitor ODQ or protein kinase C inhibitors (Ro 32-0432 and Ro 31-8220). Ischemia also stimulated caspase-3-like activity, and this was substantially reduced by pre-perfusion with DETA/NO. Reperfusion after 30 min of ischemia caused no further caspase activation, but was accompanied by necrosis, which was completely prevented by DETA/NO, and this protection was blocked by the PKG inhibitor. Incubation of isolated heart mitochondria with activated PKG blocked calcium-induced mitochondrial permeability transition and cytochrome c release. Perfusion of non-ischemic heart with DETA/NO also made the subsequently isolated mitochondria resistant to calcium-induced permeabilisation, and this protection was blocked by the PKG inhibitor. CONCLUSION: The results indicate that NO rapidly protects the ischemic heart from apoptosis and mitochondrial dysfunction via PKG-mediated blockage of mitochondrial permeability transition and cytochrome c release.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Citocromos c/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Isquemia Miocárdica/patología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/fisiología , Triazenos/farmacología , Animales , Carbazoles/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Indoles/farmacología , Masculino , Mitocondrias Cardíacas/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Ratas , Ratas Wistar
19.
Data Brief ; 13: 707-712, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28748204

RESUMEN

The data presented in this article are related to the research article entitled "Rotenone decreases ischemia-induced injury by inhibiting mitochondrial permeability transition in mature brains" (Rekuviene et al., 2017) [1]. Data in this article present the direct effects of rotenone on calcium retention capacity (CRC) in isolated normal cortex and cerebellum mitochondria, effects of rotenone intravenous infusion on leak and phosphorylating respiration rates of isolated cortex and cerebellum mitochondria, on activities of respiratory chain complexes I and II in freezed-thawed/sonicated cortex and cerebellum mitochondria after brain ischemia. In addition, detailed experimental procedures of isolation of brain mitochondria, measurements of CRC, respiration, activities of respiratory chain complexes and H2O2 generation in cortex and cerebellum mitochondria are described.

20.
Neurosci Lett ; 653: 45-50, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28527718

RESUMEN

The mitochondrial permeability transition pore (mPTP) is thought to be implicated in brain ischemia-induced cell death. Here we sought to determine whether complex I (CI) of the mitochondrial electron transfer system may be involved in regulation of mPTP opening during ischemia and whether a specific inhibitor of this complex - rotenone can protect against ischemia-induced cell death in an experimental model of total ischemia in adult rat brains. Anesthetized Wistar rats were administered a single injection of rotenone (0.01mg/kg) to the tail vein and brains were removed and subjected to 120min ischemia. We found that intravenous injection of rotenone 20min before ischemia increased resistance to Ca2+-induced mPTP opening and decreased production of reactive oxygen species (ROS) in mitochondria isolated from ischemia-damaged cortex and cerebellum. Rotenone administration before ischemia decreased infarct size in both brain regions (cortex and cerebellum). Rotenone added directly to normal, non-ischemic cortical or cerebellar mitochondria increased their resistance to Ca2+-induced mPTP opening at concentration which fully inhibited NAD-dependent mitochondrial respiration. Our data demonstrate that rotenone used intravenously may be protective against acute brain ischemia-induced injuries by inhibition of mPTP opening and ROS production. These findings suggest that CI of mitochondrial electron transfer system plays a role in mPTP regulation during cerebral ischemia in mature brains and that agents acting on CI activity may be clinically useful for stroke therapy.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Fármacos Neuroprotectores/administración & dosificación , Rotenona/administración & dosificación , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Complejo I de Transporte de Electrón/metabolismo , Masculino , Mitocondrias/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA