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.
Mol Neurobiol ; 59(5): 2977-2991, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35257284

RESUMO

Imeglimin is a novel oral antidiabetic drug modulating mitochondrial functions. However, neuroprotective effects of this drug have not been investigated. The aim of this study was to investigate effects of imeglimin against ischemia-induced brain damage and neurological deficits and whether it acted via inhibition of mitochondrial permeability transition pore (mPTP) and suppression of microglial activation. Ischemia in rats was induced by permanent middle cerebral artery occlusion (pMCAO) for 48 h. Imeglimin (135 µg/kg/day) was injected intraperitoneally immediately after pMCAO and repeated after 24 h. Immunohistochemical staining was used to evaluate total numbers of neurons, astrocytes, and microglia as well as interleukin-10 (IL-10) producing cells in brain slices. Respiration of isolated brain mitochondria was assessed using high-resolution respirometry. Assessment of ionomycin-induced mPTP opening in intact cultured primary rat neuronal, astrocytic, and microglial cells was performed using fluorescence microscopy. Treatment with imeglimin significantly decreased infarct size, brain edema, and neurological deficits after pMCAO. Moreover, imeglimin protected against pMCAO-induced neuronal loss as well as microglial proliferation and activation, and increased the number of astrocytes and the number of cells producing anti-inflammatory cytokine IL-10 in the ischemic hemisphere. Imeglimin in vitro acutely prevented mPTP opening in cultured neurons and astrocytes but not in microglial cells; however, treatment with imeglimin did not prevent ischemia-induced mitochondrial respiratory dysfunction after pMCAO. This study demonstrates that post-stroke treatment with imeglimin exerts neuroprotective effects by reducing infarct size and neuronal loss possibly via the resolution of neuroinflammation and partly via inhibition of mPTP opening in neurons and astrocytes.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Fármacos Neuroprotetores , Animais , Ratos , Encéfalo/metabolismo , Lesões Encefálicas/tratamento farmacológico , Isquemia Encefálica/complicações , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Interleucina-10/metabolismo , Mitocôndrias , Doenças Neuroinflamatórias , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Triazinas , Poro de Transição de Permeabilidade Mitocondrial
2.
J Bioenerg Biomembr ; 53(5): 499-511, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34240271

RESUMO

Recent studies have identified that under stimulation by bacterial lipopolysaccharide mammalian macrophages produce itaconic acid. Yet, it is unknown whether itaconate has any effect on viability of brain cells. Here we used extracellularly added itaconate to investigate its effects on viability of cerebellar granule cells (CGC) in cultures and respiratory functions of these cells and isolated brain mitochondria. We found that 3-5 mM itaconate had no effect on the viability of neurons, but 10 mM itaconate was toxic and induced neuronal apoptosis. Removal of itaconate after 24 h incubation resulted in further decrease in viability and number of neurons. Respiration of intact neurons was not affected by itaconate, but permeabilized cells as well as isolated brain mitochondria demonstrated decreased rates of respiration in the presence of itaconate. Using isolated adult rat brain mitochondria we found that itaconate decreased mitochondrial phosphorylating respiration, mitochondrial calcium retention capacity, production of reactive oxygen species with Complex I and Complex II substrates as well as inhibition of Complex I, Complex IV and ATP synthase. In conclusion, the results suggest that itaconic acid at millimolar concentrations affects mitochondrial functions and viability of neurons.


Assuntos
Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Succinatos/uso terapêutico , Animais , Humanos , Ratos , Espécies Reativas de Oxigênio , Succinatos/farmacologia
3.
Biomolecules ; 10(10)2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33019635

RESUMO

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.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Metformina/administração & dosagem , Mitocôndrias/efeitos dos fármacos , Fenformin/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Complexo I de Transporte de Elétrons/genética , Humanos , Masculino , Mitocôndrias/genética , Poro de Transição de Permeabilidade Mitocondrial/antagonistas & inibidores , Poro de Transição de Permeabilidade Mitocondrial/metabolismo , Necrose Dirigida por Permeabilidade Transmembrânica da Mitocôndria/efeitos dos fármacos , Necrose/tratamento farmacológico , Necrose/metabolismo , Necrose/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Fosforilação Oxidativa/efeitos dos fármacos , Ratos
4.
J Neurosci Res ; 93(3): 475-86, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25400096

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Morte Celular/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Microglia/metabolismo , Neurônios/metabolismo , Ratos
5.
BMC Gastroenterol ; 13: 159, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24229374

RESUMO

BACKGROUND: The aim of this study is to evaluate the role of NADPH oxidase in primary intestinal epithelial cells during the active phase of UC. METHODS: The primary human colonic epithelial cells were isolated from 19 patients with mild to moderate inflammatory activity of UC and 14 controls using chelation method. The cells were cultivated under the effect of mediators. Viability of cells was assessed by fluorescent microscopy. Production of reactive oxygen species (ROS) by the cells was measured fluorimetrically using Amplex Red. Production of TNF-α cytokine by the colonic epithelial cells was analysed by ELISA. RESULTS: The results of our study showed that unstimulated cells of UC patients had a decreased viability, increased ROS production, but similar TNF-α level when compared to the controls. Stimulation with LPS increased hydrogen peroxide and TNF-α level in the UC group. Treatment of colonic epithelial cells with NADPH oxidase inhibitor increased cell viability decreased the levels of ROS and TNF-α in the LPS-treated cells isolated from UC patients. CONCLUSIONS: Our study showed that bacterial endotoxins induced NADPH oxidase activation in the colonic epithelial cells. Moreover, we revealed that treatment with NADPH oxidase inhibitors had a protective effect against pro-inflammatory action of LPS in human colonic epithelium cells during inflammation.


Assuntos
Colite Ulcerativa/metabolismo , Colo/metabolismo , Células Epiteliais/metabolismo , Peróxido de Hidrogênio/metabolismo , Mucosa Intestinal/metabolismo , NADPH Oxidases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Estudos de Casos e Controles , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colite Ulcerativa/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Feminino , Humanos , Peróxido de Hidrogênio/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Lipopolissacarídeos/farmacologia , Masculino , Microscopia de Fluorescência , Pessoa de Meia-Idade , NADPH Oxidases/antagonistas & inibidores , Oniocompostos/farmacologia , Espécies Reativas de Oxigênio/imunologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/imunologia , Adulto Jovem
6.
J Neurochem ; 126(5): 604-15, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23745639

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/toxicidade , Microglia/efeitos dos fármacos , Síndromes Neurotóxicas/patologia , Animais , Anticorpos Monoclonais/imunologia , Complexo Antígeno-Anticorpo/imunologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/toxicidade , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
7.
Medicina (Kaunas) ; 47(2): 107-12, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21734444

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Ácido Egtázico/farmacologia , Endocitose/efeitos dos fármacos , Estradiol/farmacologia , Neurônios/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Microscopia de Fluorescência , Necrose , Neurônios/metabolismo , Neurônios/patologia , Propídio/farmacologia , Ratos , Ratos Wistar , beta-Ciclodextrinas/farmacologia
8.
Arch Biochem Biophys ; 496(2): 84-92, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20153288

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/toxicidade , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Peso Molecular , Ratos , Ratos Wistar , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...