Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Mol Pharmacol ; 14(2): 253-260, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32310055

RESUMO

AIMS: To show that acetate attenuates neuroinflammatory responses in activated microglia. BACKGROUND: Dietary acetate supplementation alleviates neuroglial activation in a rat model of neuroinflammation induced by intraventricular administration of lipopolysaccharide (LPS). However, the precise mechanism(s) underlying the anti-inflammatory effect of acetate, is not fully understood. OBJECTIVE: To determine whether acetate has inhibitory effects on LPS-induced neuroinflammatory responses in microglia. METHODS: We examined LPS-stimulated nitric oxide (NO) production in primary rat microglia and BV-2 cells. Protein expression of inducible NO synthase (iNOS) was determined by western blot analysis. The intracellular generation of reactive oxygen species (ROS) and glutathione (GSH) were also evaluated. RESULTS: In primary microglia, acetate decreased LPS-stimulated NO production in a dose-dependent manner, reaching significance at greater than 10 mM, and cell viability was not affected. Acetate suppressed LPS-induced expression of iNOS protein concomitantly with the decrease in NO. The LPS-induced increase in intracellular ROS production was attenuated by acetate. In addition, acetate prevented LPS-induced reduction of GSH. Notably, such suppressive effects of acetate on NO and ROS production were not observed in BV-2 cells. CONCLUSION: These findings suggest that acetate may alleviate neuroinflammatory responses by attenuating NO and ROS production in primary microglia but not in BV-2 cells. Other: All animals received humane care, and the animal protocols used in this study were approved by the Ethics Committees for Animal Experimentation.


Assuntos
Acetatos/farmacologia , Lipopolissacarídeos/metabolismo , Microglia/citologia , Doenças Neuroinflamatórias/metabolismo , Óxido Nítrico/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Glutationa/metabolismo , Humanos , Macrófagos/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
2.
Biochem Biophys Res Commun ; 526(1): 213-217, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32204914

RESUMO

The Cre-loxP recombination system is widely used to generate genetically modified mice for biomedical research. Recently, a highly efficient photoactivatable Cre (PA-Cre) based on reassembly of split Cre fragments has been established. This technology enables efficient DNA recombination that is activated upon blue light illumination with spatiotemporal precision. In this study, we generated a tTA-dependent photoactivatable Cre-loxP recombinase knock-in mouse model (TRE-PA-Cre mice) using a CRISPR/Cas9 system. These mice were crossed with ROSA26-tdTomato mice (Cre reporter mouse) to visualize DNA recombination as marked by tdTomato expression. We demonstrated that external noninvasive LED blue light illumination allows efficient DNA recombination in the liver of TRE-PA-Cre:ROSA26-tdTomato mice transfected with tTA expression vectors using hydrodynamic tail vein injection. The TRE-PA-Cre mouse established here promises to be useful for optogenetic genome engineering in a noninvasive, spatiotemporal, and cell-type specific manner in vivo.


Assuntos
Técnicas de Introdução de Genes , Engenharia Genética , Genoma , Integrases/metabolismo , Optogenética , Animais , Sequência de Bases , DNA/genética , Feminino , Luz , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais , Tetraciclina/farmacologia
3.
Commun Biol ; 2: 346, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552299

RESUMO

The pathophysiological role of mammalian target of rapamycin complex 1 (mTORC1) in neurodegenerative diseases is established, but possible therapeutic targets responsible for its activation in neurons must be explored. Here we identified solute carrier family 38a member 1 (SNAT1, Slc38a1) as a positive regulator of mTORC1 in neurons. Slc38a1flox/flox and Synapsin I-Cre mice were crossed to generate mutant mice in which Slc38a1 was selectively deleted in neurons. Measurement of 2,3,5-triphenyltetrazolium chloride (TTC) or the MAP2-negative area in a mouse model of middle cerebral artery occlusion (MCAO) revealed that Slc38a1 deficiency decreased infarct size. We found a transient increase in the phosphorylation of p70S6k1 (pp70S6k1) and a suppressive effect of rapamycin on infarct size in MCAO mice. Autophagy inhibitors completely mitigated the suppressive effect of SNAT1 deficiency on neuronal cell death under in vitro stroke culture conditions. These results demonstrate that SNAT1 promoted ischemic brain damage via mTOR-autophagy system.


Assuntos
Sistema A de Transporte de Aminoácidos/antagonistas & inibidores , Sistema A de Transporte de Aminoácidos/metabolismo , Autofagia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Sistema A de Transporte de Aminoácidos/genética , Animais , Infarto Cerebral/etiologia , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Expressão Gênica , Loci Gênicos , Genoma , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuroproteção , Especificidade de Órgãos
4.
Nihon Yakurigaku Zasshi ; 153(2): 67-72, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-30745516

RESUMO

Mesenchymal stem cell (MSC) is a type of tissue stem cell. In clinical studies, cultured MSCs have shown important therapeutic effects on diseases via the reduction of neurological defects and regulation of immune responses. However, in vivo MSC localization, function, and properties are poorly understood; therefore, the molecular understanding of MSCs hierarchy is less advanced compared to hematopoietic stem cell hierarchy. To address these issues, we developed a method that enables us to visualize MSCs, manipulate their function, and analyze their molecular biology in vivo. Paired-related homeobox 1 (Prrx1)-positive cells are transiently observed during limb skeletal development in mice. Prrx1-positive cells form heterogeneous populations comprising multiple mesenchymal progenitors with different lineages that are developing into osteoblasts, chondrocytes, adipocytes, fibroblasts, and tendon and ligament cells. Our results suggest that osteoblast differentiation in the calvaria begins at the Prrx1+Sca1+ MSC stage with sequential progression to Prrx1+Sca1- cells, then Osterix+Prrx1-Sca1- osteoblast precursors, which eventually form mature α1(I)-collagen+ osteoblasts. Using Runt-related transcription factor 2 (Runx2) conditional knockout mice, furthermore, we found that the essential period of Runx2 function in intramembranous ossification likely begins at the Prrx1+Sca1+ MSC stage and ends at the Osterix+Prrx1-Sca1- osteoblast precursor stage (before mature the α1(I)-collagen+ osteoblasts appear). This approach will enable us to understand the in vivo molecular biology features of MSCs, leading to their therapeutic applications for tissue repair and regeneration. This development can also contribute to the field of pluripotent stem cell by enabling the transplantation of lineage-restricted mesenchymal progenitors.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteogênese , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Knockout
5.
Biochem Biophys Res Commun ; 509(4): 1028-1033, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30660360

RESUMO

Runt-related transcription factor 2 (Runx2), also known as core binding factor 1 (Cbfa1), is a multifunctional transcription factor and an essential master gene controlling osteoblast differentiation. We previously demonstrated the in vivo functions of Runx2 in mesoderm-derived cells. However, no studies have been conducted on Runx2 function during the differentiation of neural crest (NC)-derived cells in vivo. Wingless-type MMTV integration site family member 1 (Wnt1) is expressed in the NC, and Wnt1-Cre efficiently targets craniofacial NC-derived cells. Runx2 deficiency in cells of the Wnt1 lineage (referred henceforth as Runx2wnt1-/- within mice) resulted in defective ossification in certain regions, primarily in the anterior half of the craniofacial bones, including the frontal bone, jugal bone, squamous temporal bone, mandible, maxilla, and nasal bone. The skeletal analysis also revealed that heterozygous Runx2wnt1+/- embryos had an impaired closure of the frontal bone at the metopic suture and lacked the secondary palate in spite of otherwise normal ossification. This result suggests that ossification at the central part of the frontal bone is more dependent on Runx2 expression in comparison to other areas. These results indicate that Runx2 is indispensable not only for mesoderm-derived cells but also for NC-derived cells to differentiate during intramembranous ossification after migration to their destination from the neural plate border. Moreover, this implies that there are different levels of dependency on Runx2 expression for successful ossification between NC-derived cells that have migrated to different locations.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Crista Neural/citologia , Osteogênese , Animais , Diferenciação Celular , Movimento Celular , Anormalidades Craniofaciais/etiologia , Embrião de Mamíferos , Camundongos , Crista Neural/embriologia , Proteína Wnt1/metabolismo
6.
Neurochem Res ; 43(11): 2047-2054, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30203400

RESUMO

Glioblastoma (GBM) is the most aggressive and lethal form of brain tumor. However, therapeutic strategies against malignant gliomas have not been completely established. Runt-related transcription factor 2 (Runx2) is an essential gene for skeletal development but its regulatory role in the malignant progression of glioma remains unclear. Here we investigated expression levels of RUNX2 in glioma tissues and its regulatory effects on aberrant growth of glioma cells. RUNX2 mRNA levels were higher in GBM tissues than that of normal brains or low-grade gliomas. RUNX2 protein was detected in five out of seven human GBM cell lines and its level was positively correlated with proliferative capacity. Stable transduction of dominant-negative Runx2 in rat-derived C6 glioma cells not only inhibited the promoter activity containing Runx2 response element, but also decreased mRNA expression levels of Runx2 target genes, such as Mmp13 and Spp1, as well as the proliferative capacity. Furthermore, transient introduction of Runx2-targeted siRNAs into C6 glioma cells significantly decreased mRNA expression levels of Mmp13 and Spp1 and the proliferative capacity. Furthermore, Runx2 knockdown suppressed both Ccnd1 mRNA expression and activation of the Ccnd1 promoter by forskolin, a PKA-activating reagent, in C6 glioma cells. Our results demonstrate that cross-talk between cAMP/PKA signaling and RUNX2 promotes a malignant phenotype of glioma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glioma/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Glioma/genética , Humanos , Ratos
7.
Neurochem Res ; 43(9): 1723-1735, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29947014

RESUMO

In several neurodegenerative diseases such as Alzheimer's disease (AD), microglia are hyperactivated and release nitric oxide (NO) and proinflammatory cytokines, resulting its neuropathology. Mounting evidence indicates that dietary supplementation with coconut oil (CNO) reduces the cognitive deficits associated with AD; however, the precise mechanism(s) underlying the beneficial effect of CNO are unknown. In the present study, we examined the effects of lauric acid (LA), a major constituent of CNO, on microglia activated experimentally by lipopolysaccharide (LPS), using primary cultured rat microglia and the mouse microglial cell line, BV-2. LA attenuated LPS-stimulated NO production and the expression of inducible NO synthase protein without affecting cell viability. In addition, LA suppressed LPS-induced reactive oxygen species and proinflammatory cytokine production, as well as phosphorylation of p38-mitogen activated protein kinase and c-Jun N-terminal kinase. LA-induced suppression of NO production was partially but significantly reversed in the presence of GW1100, an antagonist of G protein-coupled receptor (GPR) 40, which is an LA receptor on the plasma membrane. LA also decreased LPS-induced phagocytosis, which was completely reversed by co-treatment with GW1100. Moreover, LA alleviated amyloid-ß-induced enhancement of phagocytosis. These results suggest that attenuation of microglial activation by LA may occur via the GPR40-dependent pathway. Such effects of LA may reduce glial activation and the subsequent neuronal damage in AD patients who consume CNO.


Assuntos
Ácidos Láuricos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Animais , Linhagem Celular , Óleo de Coco/farmacologia , Citocinas/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fosforilação/efeitos dos fármacos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
8.
Int J Inflam ; 2018: 8496973, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29692883

RESUMO

Cumulative evidence indicates that estrogen receptor (ER) agonists attenuate neuroinflammation. Equol, a major isoflavone from soybean, exhibits estrogen-like biological activity, but their effect on inflammatory response has not been well established. Here, we investigated the effect of S-equol on nitric oxide (NO) production, well-known inflammatory change in astrocytes stimulated by LPS. S-Equol attenuated LPS-induced NO production with a concomitant decrease in expression of inducible NO synthase (iNOS). S-Equol did not affect LPS-induced increase in intracellular ROS production. Intracellular ER blocker ICI 182.780 had no effect on S-equol-induced decrease in NO production. Addition of G-15, antagonist of G protein-coupled receptor 30 which is nongenomic ER and located on cell surface, partially recovered S-equol-induced attenuation of NO production. These findings suggest that attenuation of NO production by S-equol may mitigate LPS-induced neuroinflammation in astrocytes. S-Equol may exert a glioprotective effect, at least in part, via a nongenomic effect.

9.
Neurochem Res ; 43(1): 41-49, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28466190

RESUMO

Activation of glial cells has been observed in neurodegenerative diseases including Alzheimer's disease (AD). Aggregation of amyloid ß (Aß) is profusely observed as characteristic pathology in AD brain. In our previous study using microglial cell line BV-2, tissue-type transglutaminase (TG2) was found to be involved in phagocytosis (Kawabe et al., in Neuroimmunomodulation 22(4):243-249, 2015; Kawabe et al., Neurochem Res 2017). In the present study, we examined whether TG2 and milk fat globule EGF factor 8 protein (MFG-E8), an adaptor protein promotes macrophage to engulf apoptotic cells, were involved in Aß endocytosis. When the neuronal/glial mixed culture was stimulated freshly prepared Aß1-42 for 3 days, the incorporation of Aß was observed by immunofluorescence staining technique in Iba-1-positive microglia. Cystamine, a broad competitive inhibitor of TGs, suppressed it. When aggregated Aß was added to the mixed culture, the immunoreactivity of MFG-E8 surrounding Aß was observed, and then followed by microglial endocytosis. Using western blotting technique, MFG-E8 was detected in cell lysate of astrocyte culture, and was also detected in the medium. When microglia culture was incubated with astrocyte conditioned medium, MFG-E8 levels in microglia tended to increase. It is likely that microglia might utilize MFG-E8 released from astrocytes as well as that expressed in themselves in order to endocytose Aß aggregation. Furthermore, we confirmed that MFG-E8 could bind with TG2 in microglia culture by immunoprecipitate technique. These results suggest that microglia might uptake Aß as a complex of aggregated Aß/MFG-E8/TG2.


Assuntos
Antígenos de Superfície/metabolismo , Endocitose/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Microglia/metabolismo , Proteínas do Leite/metabolismo , Transglutaminases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Células Cultivadas , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Gotículas Lipídicas , Neurônios/metabolismo , Fagocitose/fisiologia , Proteína 2 Glutamina gama-Glutamiltransferase , Ratos
10.
Neurochem Res ; 43(2): 363-374, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29124566

RESUMO

Zn2+ plays a crucial role in the CNS where it accumulates in synaptic vesicles and is released during neurotransmission. Synaptically released Zn2+ is taken up by neurons and astrocytes. The majority of previous work has focused on neuronal damage caused by excess Zn2+. However, its effect on astrocyte function is not well understood. We examined the effect of extracellularly applied Zn2+ on nitric oxide (NO) production in primary cultured rat astrocytes, which were experimentally activated by lipopolysaccharide (LPS). Zn2+, at a concentration up to 125 µM, augmented LPS-induced NO production without affecting cell viability. LPS induced expression of both mRNA and protein of inducible NO synthase; this expression was enhanced by 125 µM Zn2+. Zn2+ also increased LPS-induced production of intracellular reactive oxygen species. Zn2+ enhanced the phosphorylation of p38-mitogen-activated protein kinase (MAPK) at 1-6 h after LPS treatment. The LPS-induced nuclear factor-kappaB (NFκB) activation was sustained for 6 h by Zn2+. Intracellular Zn2+ chelation with N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) or inhibition of p38-MAPK diminished the Zn2+ enhancement of LPS-induced NO production. These findings suggest that activation of MAPK and NFκB is important for mediating Zn2+enhancement of LPS-induced NO production in astrocytes. Such changes may exacerbate glial and neuronal damage during neuroinflammation.


Assuntos
Astrócitos/metabolismo , Lipopolissacarídeos/farmacologia , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Zinco/metabolismo , Animais , Células Cultivadas , Microglia/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuroglia/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
11.
Neurochem Int ; 119: 171-177, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29108865

RESUMO

Insulin resistance in brain has been reported in Alzheimer's diseases (AD). Insulin signaling is important for homeostasis in brain function and reported to be disturbed in neurons leading to tau phosphorylation and neurofibrillary tangles. Many investigations of insulin in neurons have been reported; however, it has not been reported whether astrocytes also produce insulin. In the present study, we assessed the expression of insulin in astrocytes cultured from rat embryonic brain and the effects of amyloid ß1-42 (Aß) and lipopolysaccharide (LPS) on the expression. We found that astrocytes expressed preproinsulin mRNAs and insulin protein, and that Aß or LPS decreased these expressions. Antioxidants, glutathione and N-acetylcysteine, restored the decreases in insulin mRNA expression by Aß and by LPS. Insulin protein was detected in astrocyte conditioned medium. These results suggest that astrocytes express and secrete insulin. Oxidative stress might be involved in the decreased insulin expression by Aß or LPS. The insulin decrease by Aß in astrocytes could be a novel disturbing mechanism for brain insulin signaling in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Antioxidantes/farmacologia , Astrócitos/metabolismo , Insulina/metabolismo , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Precursores de Proteínas/metabolismo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/metabolismo
12.
Neurochem Int ; 119: 126-131, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-28390951

RESUMO

Microglial activation has been suggested to play important roles in various neurodegenerative diseases by phagocytosis and producing various factors such as nitric oxide (NO), proinflammatory cytokines. Excessive production of NO, as a consequence of increased inducible nitric oxide synthase (iNOS) in microglia, contributes to the neurodegeneration. During a search for compounds that regulate endoplasmic reticulum (ER) stress, a dibenzoylmethane derivative, 2,2'-dimethoxydibenzoylmethane (DBM 14-26) was identified as a novel neuroprotective agent (Takano et al., Am. J. Physiol. Cell Physiol. 293, C1884-1894, 2007). We previously reported in cultured astrocytes that DBM 14-26 protected hydrogen peroxide-induced cell death and inhibited lipopolysaccharide (LPS)-induced NO production (Takano et al., J. Neurosci. Res. 89, 955-965, 2011). In the present study, we assessed the effects of DBM 14-26 on microglia using the mouse cell line BV-2 and found that DBM 14-26 inhibited LPS-induced iNOS expression and NO production also in microglia. DBM 14-26 also suppressed LPS-induced IL-1ß expression. Conditioned medium of BV-2 cells stimulated by LPS significantly decreased cell viability of neuron (human neuroblastoma SH-SY5Y cells) compared with the absence of LPS. Conditioned medium of BV-2 cells stimulated by LPS in the presence of DBM 14-26 did not significantly decreased cell viability of neuron. These results indicate that microglial activation by LPS causes neuronal cell death and DBM 14-26 protect neuron through the inhibition of microglial activation. Functional regulation of microglia by DBM 14-26 could be a therapeutic candidate for the treatment of neurodegenerative diseases.


Assuntos
Astrócitos/efeitos dos fármacos , Chalconas/farmacologia , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Linhagem Celular , Citocinas/metabolismo , Lipopolissacarídeos/farmacologia , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Neurochem Res ; 43(1): 59-65, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28589517

RESUMO

Glutamate uptake is a main function of astrocytes to keep extracellular glutamate levels low and protect neurons against glutamate-induced excitotoxicity. On the other hand, astrocyte networks formed by gap junctions, which are consisted with connexins and connecting neighboring cells, are reported to play a critical role in maintaining the homeostasis in the brain. In the present study, we examined the effects of gap junction inhibitors on the glutamate uptake activity in cultured rat cortical astrocytes. At first, we confirmed the effects of gap junction inhibitors, 1-octanol and carbenoxolone, on cell-cell communication by the scrape-loading assay using a fluorescent dye Lucifer yellow. Both of 1-octanol and carbenoxolone treatments for 20 min in cultured astrocytes significantly suppressed the cell-cell communication assessed as the distance of dye-spreading. 1-octanol and carbenoxolone increased the glutamate uptake by astrocytes and glutamate aspartate transporter (GLAST) expression on the cell membrane. These results suggest that gap junction inhibitors increase the glutamate uptake activity through the increase of GLAST proteins located on the cell membrane. The regulation of gap junction in astrocytes might protect neurons against glutamate-induced excitotoxicity.


Assuntos
Astrócitos/metabolismo , Junções Comunicantes/metabolismo , Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Transporte Biológico/fisiologia , Comunicação Celular/fisiologia , Células Cultivadas , Conexinas/metabolismo , Ratos Wistar
14.
J Neurosci ; 37(42): 10052-10062, 2017 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-28912161

RESUMO

Circadian rhythm disturbances are well established in neurological diseases. However, how these disruptions cause homeostatic imbalances remains poorly understood. Brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1) is a major circadian clock transcriptional activator, and Bmal1 deficiency in male Bmal1nestin-/- mice induced marked astroglial activation without affecting the number of astrocytes in the brain and spinal cord. Bmal1 deletion caused blood-brain barrier (BBB) hyperpermeability with an age-dependent loss of pericyte coverage of blood vessels in the brain. Using Nestin-green fluorescent protein (GFP) transgenic mice, we determined that pericytes are Nestin-GFP+ in the adult brain. Bmal1 deletion caused Nestin-GFP+ pericyte dysfunction, including the downregulation of platelet-derived growth factor receptor ß (PDGFRß), a protein necessary for maintaining BBB integrity. Knockdown of Bmal1 downregulated PDGFRß transcription in the brain pericyte cell line. Thus, the circadian clock component Bmal1 maintains BBB integrity via regulating pericytes.SIGNIFICANCE STATEMENT Circadian rhythm disturbances may play a role in neurodegenerative disorders, such as Alzheimer's disease. Our results revealed that one of the circadian clock components maintains the integrity of the blood-brain barrier (BBB) by regulating vascular-embedded pericytes. These cells were recently identified as a vital component for the control of BBB permeability and cerebral blood flow. Our present study demonstrates the involvement of circadian clock component Bmal1 in BBB homeostasis and highlights the role of Bmal1 dysfunction in multiple neurological diseases.


Assuntos
Fatores de Transcrição ARNTL/deficiência , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Pericitos/metabolismo , Pericitos/patologia , Fatores de Transcrição ARNTL/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Linhagem Celular , Ritmo Circadiano/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
15.
Neurochem Res ; 42(8): 2384-2391, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28391388

RESUMO

Activation of astrocytes has been observed in neurodegenerative diseases including Alzheimer's disease (AD). Transglutaminase (TG) is a crosslinking enzyme and contributes to cell adhesion, cytoskeleton construct, extracellular matrix formation, and so on. One of the isozymes, tissue-type TG (TG2) is reported to be activated in AD. Moreover, amyloid ß1-42 (Aß), which is aggregated and the aggregation is detected as characteristic pathology in AD brain, is known to be a substrate of TG2. However, contribution and derivation of TGs in brain for Aß aggregation remain to be clarified. In the present study, we examined the effects of cultured astrocytes prepared from rat embryonic brain cortex on Aß aggregation. When freshly prepared Aß was added to cultured astrocytes for 7 days, Aß monomer decreased and Aß oligomer unchanged. On the other hand, when Aß monomer was diluted with astrocytes conditioned medium, Aß oligomer increased time-dependently, and an inhibitor of TGs, cystamine, blocked it. Furthermore, when cultured astrocytes were stimulated with aggregated Aß, TG2 expression significantly increased. These results suggest that astrocytes could uptake Aß monomer to eliminate from brain; however, TGs derived from astrocytes might accelerate Aß aggregation and the aggregated Aß might enhance TG2 in astrocytes as a vicious cycle in pathological conditions. Adequate control of TGs expression and function in astrocytes would be an important factor in AD pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Transglutaminases/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Células Cultivadas , Feminino , Fragmentos de Peptídeos/farmacologia , Gravidez , Proteína 2 Glutamina gama-Glutamiltransferase , Ratos , Ratos Wistar , Transglutaminases/isolamento & purificação
16.
Neurochem Res ; 42(5): 1488-1495, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28224343

RESUMO

Amphotericin B (AmB), a polyene antibiotic, is reported to cause the microglial activation to induce nitric oxide (NO) production and proinflammatory cytokines expression, and change neurotrophic factors expression in cultured microglia (Motoyoshi et al. in Neurochem Int 52:1290-1296, 2008). On the other hand, tissue-type transglutaminase (TG2) is involved in connection to phagocytes with apoptotic cells. Engulfment of neurons by activated microglia is thought to cause neurodegenerative diseases but detail is unclear, and involvement of TG2 in phagocytosis has been reported in our previous study using lipopolysaccharide-stimulated BV-2 cells (Kawabe et al. in Neuroimmunomodulation 22(4):243-249, 2015). In the present study, we examined the changes of TG2 expression, phagocytosis and pinocytosis in BV-2 cells stimulated by AmB. AmB stimulation increased TG2 expression and TG activity. Phagocytosis of dead cells and pinocytosis of fluorescent microbeads were also up-regulated by AmB stimulation in BV-2 cells. Blockade of TG activity by cystamine, an inhibitor of TGs, suppressed AmB-enhanced TG2 expression, TG activity, NO production, phagocytosis and pinocytosis. Excessive NO production from microglia and/or facilitation of phagocytosis might be involved in neuronal death. To control TG activity might make possible to protect neurons and care for CNS diseases.


Assuntos
Anfotericina B/farmacologia , Endocitose/fisiologia , Proteínas de Ligação ao GTP/biossíntese , Regulação Enzimológica da Expressão Gênica , Microglia/enzimologia , Transglutaminases/biossíntese , Regulação para Cima/fisiologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Endocitose/efeitos dos fármacos , Proteínas de Ligação ao GTP/genética , Camundongos , Microglia/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/genética , Regulação para Cima/efeitos dos fármacos
17.
Neurochem Res ; 41(11): 3138-3146, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27542961

RESUMO

The biomolecule acetate can be utilized for energy production, lipid synthesis, and several metabolic processes. Acetate supplementation reduces neuroglial activation in a model of neuroinflammation induced by intraventricular injection of lipopolysaccharide (LPS). To investigate the mechanisms underlying the anti-inflammatory effect of acetate on glial cells, we examined the effect of acetate on nitric oxide (NO) production, which was experimentally activated by LPS, in cultured primary rat astrocytes. Acetate attenuated the LPS-induced NO production in a dose-dependent manner, although cell viability was not affected. Acetate suppressed the phosphorylation of p38-mitogen-activated protein kinase 24 h after LPS treatment. Acetate decreased the LPS-induced production of intracellular reactive oxygen species (ROS) at 4-24 h concomitant with an increase in glutathione. Acetate rescued astrocytes from the hydrogen peroxide-induced cell death by reducing ROS levels. These findings suggest that attenuation of NO production by acetate may alleviate glial cell damage during neuroinflammation. Acetate may offer a glioprotective effect through an anti-oxidative mechanism.


Assuntos
Astrócitos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Óxido Nítrico/biossíntese , Animais , Astrócitos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Inflamação/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuroglia/metabolismo , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
18.
Biochim Biophys Acta ; 1852(9): 1787-95, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25952905

RESUMO

The mitochondrial aspartate-glutamate carrier isoform 2 (citrin) and mitochondrial glycerol-3-phosphate dehydrogenase (mGPD) double-knockout mouse has been a useful model of human citrin deficiency. One of the most prominent findings has been markedly increased hepatic glycerol 3-phosphate (G3P) following oral administration of a sucrose solution. We aimed to investigate whether this change is detectable outside of the liver, and to explore the mechanism underlying the increased hepatic G3P in these mice. We measured G3P and its metabolite glycerol in plasma and urine of the mice under various conditions. Glycerol synthesis from fructose was also studied using the liver perfusion system. The citrin/mGPD double-knockout mice showed increased urine G3P and glycerol under normal, fed conditions. We also found increased plasma glycerol under fasted conditions, while oral administration of different carbohydrates or ethanol led to substantially increased plasma glycerol. Fructose infusion to the perfused liver of the double-knockout mice augmented hepatic glycerol synthesis, and was accompanied by a concomitant increase in the lactate/pyruvate (L/P) ratio. Co-infusion of either pyruvate or phenazine methosulfate, a cytosolic oxidant, with fructose corrected the high L/P ratio, leading to reduced glycerol synthesis. Overall, these findings suggest that hepatic glycerol synthesis is cytosolic NADH/NAD(+) ratio-dependent and reveal a likely regulatory mechanism for hepatic glycerol synthesis following a high carbohydrate load in citrin-deficient patients. Therefore, urine G3P and glycerol may represent potential diagnostic markers for human citrin deficiency.

19.
Neuroimmunomodulation ; 22(4): 243-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25301694

RESUMO

OBJECTIVES: In peripheral macrophages, tissue-type transglutaminase (TG2) is reported to be involved in phagocytosis of apoptotic cells. However, the contribution of TG2 to microglial phagocytosis has not been investigated. In this study, using a microglial cell line, BV-2, we examined the changes in TG2 expression, phagocytosis and pinocytosis in cells stimulated by lipopolysaccharide (LPS). METHODS: Cells of the mouse microglial cell line BV-2 were stimulated by LPS with or without cystamine, an inhibitor of TG enzyme activity, for 24 h. TG2 expression was measured by real-time RT-PCR and Western blotting. TG activity was evaluated using biotinylated pentylamine as a substrate. Pinocytosis was determined by uptake of 1-µm fluorescent microbeads. Phagocytosis was assessed by uptake of dead cells, human neuroblastoma SH-SY5Y cells, which were pretreated with H2O2 for 24 h. RESULTS: Phagocytosis of dead cells and pinocytosis of fluorescent microbeads were up-regulated by LPS stimulation together with TG2 expression. Blockade of TG enzyme activity by cystamine suppressed TG2 expression, phagocytosis and pinocytosis. CONCLUSIONS: These results suggested that LPS-induced TG2 was involved in the mechanism of pinocytosis and phagocytosis in microglia.


Assuntos
Endocitose/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Lipopolissacarídeos/farmacologia , Microglia/metabolismo , Transglutaminases/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Proteína 2 Glutamina gama-Glutamiltransferase
20.
J Vet Med Sci ; 76(10): 1353-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25283947

RESUMO

Amphotericin B (AmB) is a polyene antifungal drug and is reported to be one of a few reagents having therapeutic effects on prion diseases, that is, a delay in the appearance of clinical signs and prolongation of the survival time in an animal model. In prion diseases, glial cells have been suggested to play important roles; however, the therapeutic mechanism of AmB on prion diseases remains elusive. We have previously reported that AmB changed the expression of neurotrophic factors in microglia and astrocytes (Motoyoshi et al., 2008, Neurochem. Int. 52, 1290-1296; Motoyoshi-Yamashiro et al., 2013, ibid. 63, 93-100). These results suggested that neurotrophic factors derived from glial cells might be involved in the therapeutic mechanism of AmB. In the present study, we examined immunohistochemically the effects of AmB on the expression of neurotrophic factors in the rat brain. We found that direct injection of AmB into the striatum significantly enhanced the expression of glial cell line-derived neurotrophic factor protein. Amphotericin B also increased the expressions of CD11b and glial fibrillary acidic protein, markers of microglia and astrocytes, respectively. Moreover, expressions of the two neurotrophic factors by AmB were co-localized with the expression of CD11b or glial fibrillary acidic protein. These results suggest that AmB in vivo might also activate glial cells and induce the production of neurotrophic factors protecting neurons in prion diseases.


Assuntos
Anfotericina B/farmacologia , Antifúngicos/farmacologia , Encéfalo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Encéfalo/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Microglia/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...