Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neuroinflammation ; 20(1): 11, 2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36650518

RESUMO

BACKGROUND: Ischemic stroke in white matter of the brain induces not only demyelination, but also neuroinflammation. Peripheral T lymphocytes, especially regulatory T cells (Tregs), are known to infiltrate into ischemic brain and play a crucial role in modulation of inflammatory response there. We previously reported that transplantation of vascular endothelial cells generated from human induced pluripotent stem cells (iVECs) ameliorated white matter infarct. The aim of this study is to investigate contribution of the immune system, especially Tregs, to the mechanism whereby iVEC transplantation ameliorates white matter infarct. METHODS: iVECs and human Tregs were transplanted into the site of white matter lesion seven days after induction of ischemia. The egress of T lymphocytes from lymph nodes was sequestered by treating the animals with fingolimod (FTY720). The infarct size was evaluated by magnetic resonance imaging. Immunohistochemistry was performed to detect the activated microglia and macrophages, T cells, Tregs, and oligodendrocyte lineage cells. Remyelination was examined by Luxol fast blue staining. RESULTS: iVEC transplantation reduced ED-1+ inflammatory cells and CD4+ T cells, while increased Tregs in the white matter infarct. Treatment of the animals with FTY720 suppressed neuroinflammation and reduced the number of both CD4+ T cells and Tregs in the lesion, suggesting the importance of infiltration of these peripheral immune cells into the lesion in aggravation of neuroinflammation. Suppression of neuroinflammation by FTY720 per se, however, did not promote remyelination in the infarct. FTY720 treatment negated the increase in the number of Tregs by iVEC transplantation in the infarct, and attenuated remyelination promoted by transplanted iVECs, while it did not affect the number of oligodendrocyte lineage cells increased by iVEC transplantation. Transplantation of Tregs together with iVECs into FTY720-treated ischemic white matter did not affect the number of oligodendrocyte lineage cells, while it remarkably promoted myelin regeneration. CONCLUSIONS: iVEC transplantation suppresses neuroinflammation, but suppression of neuroinflammation per se does not promote remyelination. Recruitment of Tregs by transplanted iVECs contributes significantly to promotion of remyelination in the injured white matter.


Assuntos
Células-Tronco Pluripotentes Induzidas , Substância Branca , Animais , Humanos , Substância Branca/patologia , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Linfócitos T Reguladores , Células Endoteliais , Doenças Neuroinflamatórias , Encéfalo/patologia , Isquemia/patologia , Infarto
2.
Mol Brain ; 14(1): 52, 2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33712038

RESUMO

The HapMap Project is a major international research effort to construct a resource to facilitate the discovery of relationships between human genetic variations and health and disease. The Ser19Stop single nucleotide polymorphism (SNP) of human phytanoyl-CoA hydroxylase-interacting protein-like (PHYHIPL) gene was detected in HapMap project and registered in the dbSNP. PHYHIPL gene expression is altered in global ischemia and glioblastoma multiforme. However, the function of PHYHIPL is unknown. We generated PHYHIPL Ser19Stop knock-in mice and found that PHYHIPL impacts the morphology of cerebellar Purkinje cells (PCs), the innervation of climbing fibers to PCs, the inhibitory inputs to PCs from molecular layer interneurons, and motor learning ability. Thus, the Ser19Stop SNP of the PHYHIPL gene may be associated with cerebellum-related diseases.


Assuntos
Cerebelo/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Polimorfismo de Nucleotídeo Único , Células de Purkinje/ultraestrutura , Sequência de Aminoácidos , Animais , Sistemas CRISPR-Cas , Forma Celular , Códon de Terminação , Feminino , Técnicas de Introdução de Genes , Projeto HapMap , Humanos , Interneurônios/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Aprendizagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Fibras Nervosas/fisiologia , Células de Purkinje/metabolismo , Teste de Desempenho do Rota-Rod , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
3.
J Neurochem ; 153(6): 759-771, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31883380

RESUMO

White matter infarct induces demyelination and brain dysfunction. We previously reported that transplantation of brain microvascular endothelial cells improved the behavioral outcome and promoted remyelination by increasing the number of oligodendrocyte precursor cells in the rat model of white matter infarct. In this study, we investigated the effects of transplantation of vascular endothelial cells generated from human induced pluripotent stem cells (iPSCs) on the rat model of white matter infarct. Seven days after induction of ischemic demyelinating lesion by injection of endothelin-1 into the internal capsule of a rat brain, iPSC-derived vascular endothelial cells (iVECs) were transplanted into the site of demyelination. The majority of iVECs transplanted into the internal capsule survived for 14 days after transplantation when traced by immunohistochemistry for a human cytoplasmic protein. iVEC transplantation significantly recovered hind limb rotation angle as compared to human iPSC or rat meningeal cell transplantation when evaluated using footprint test. Fourteen days after iVEC transplantation, the infarct area remarkably decreased as compared to that just before the transplantation when evaluated using magnetic resonance imaging or luxol fast blue staining, and remyelination was promoted dramatically in the infarct when assessed using luxol fast blue staining. Transplantation of iVECs increased the number of oligodendrocyte lineage cells and suppressed the inflammatory response and reactive astrocytogenesis. These results suggest that iVEC transplantation may prove useful in treatment for white matter infarct.


Assuntos
Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/terapia , Células Endoteliais/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco/métodos , Substância Branca/diagnóstico por imagem , Animais , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
4.
J Neurosci ; 39(32): 6339-6353, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31201232

RESUMO

ADP-ribosylation factors (ARFs) are a family of small monomeric GTPases comprising six members categorized into three classes: class I (ARF1, 2, and 3), class II (ARF4 and 5), and class III (ARF6). In contrast to class I and III ARFs, which are the key regulators in vesicular membrane trafficking, the cellular function of class II ARFs remains unclear. In the present study, we generated class II ARF-deficient mice and found that ARF4+/-/ARF5-/- mice exhibited essential tremor (ET)-like behaviors. In vivo electrophysiological recordings revealed that ARF4+/-/ARF5-/- mice of both sexes exhibited abnormal brain activity when moving, raising the possibility of abnormal cerebellar excitability. Slice patch-clamp experiments demonstrated the reduced excitability of the cerebellar Purkinje cells (PCs) in ARF4+/-/ARF5-/- mice. Immunohistochemical and electrophysiological analyses revealed a severe and selective decrease of pore-forming voltage-dependent Na+ channel subunit Nav1.6, important for maintaining repetitive action potential firing, in the axon initial segment (AIS) of PCs. Importantly, this decrease in Nav1.6 protein localized in the AIS and the consequent tremors in ARF4+/-/ARF5-/- mice could be alleviated by the PC-specific expression of ARF5 using adeno-associated virus vectors. Together, our data demonstrate that the decreased expression of the class II ARF proteins in ARF4+/-/ARF5-/- mice, leading to a haploinsufficiency of ARF4 in the absence of ARF5, impairs the localization of Nav1.6 to the AIS and hence reduces the membrane excitability in PCs, resulting in the ET-like movement disorder. We suggest that class II ARFs function in localizing specific proteins, such as Nav1.6, to the AIS.SIGNIFICANCE STATEMENT We found that decreasing the expression of class II ARF proteins, through the generation of ARF4+/-/ARF5-/- mice, impairs Nav1.6 distribution to the axon initial segment (AIS) of cerebellar Purkinje cells (PCs), thereby resulting in the impairment of action potential firing of PCs. The ARF4+/-/ARF5-/- mutant mice exhibited movement-associated essential tremor (ET)-like behavior with pharmacological profiles similar to those in ET patients. The exogenous expression of ARF5 reduced the tremor phenotype and restored the localization of Nav1.6 immunoreactivity to the AIS in ARF4+/-/ARF5-/- mice. Thus, our results suggest that class II ARFs are involved in the localization of Nav1.6 to the AISs in cerebellar PCs and that the reduction of class II ARF activity leads to ET-like movement disorder.


Assuntos
Fatores de Ribosilação do ADP/fisiologia , Axônios/metabolismo , Transtornos dos Movimentos/etiologia , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Células de Purkinje/metabolismo , Tremor/etiologia , Fatores de Ribosilação do ADP/deficiência , Fatores de Ribosilação do ADP/genética , Potenciais de Ação , Animais , Dependovirus/genética , Eletroencefalografia , Eletromiografia , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Genótipo , Movimentos da Cabeça , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos dos Movimentos/metabolismo , Transtornos dos Movimentos/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.6/deficiência , Técnicas de Patch-Clamp , Transporte Proteico , Células de Purkinje/fisiologia , Teste de Desempenho do Rota-Rod , Método Simples-Cego , Tremor/metabolismo , Tremor/fisiopatologia
5.
Neurosci Lett ; 692: 70-76, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30389418

RESUMO

Subcortical white matter infarction causes ischemic demyelination and loss of brain functions, as the result of disturbances of the blood flow. Although angiogenesis is one of the recovery processes after cerebral infarction, the dynamics of revascularization after white matter infarction still remains unclear. We induced white matter infarction in the internal capsule of Flk1-GFP::Flt1-tdsRed double transgenic mice by injection of endothelin-1 (ET-1), a vasoconstrictor peptide, together with N(G)-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, and followed the changes in Flk1 and Flt1 expression in the vascular system in the infarct area. Reduction of Flt1-tdsRed-positive blood vessels 1 day after the injection and increase of Flk1-GFP-strongly-positive blood vessels 3 days after the injection were apparent. PDGFRß-strongly-positive (PDGFRß+) cells appeared in the infarct area 3 days after the injection and increased their number thereafter. Three days after the injection, most of these cells were in close contact with Flk1-GFP-positive endothelial cells, indicating these cells are bona fide pericytes. Seven days after the injection, the number of PDGFRß+ cells increased dramatically, and the vast majority of these cells were not in close contact with Flk1-GFP-positive endothelial cells. Taken together, our results suggest revascularization begins early after the ischemic insult, and the emerging pericytes first ensheath blood vessels and then produce fibroblast-like cells not directly associated with blood vessels.


Assuntos
Infarto Encefálico/fisiopatologia , Neovascularização Fisiológica , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Substância Branca/irrigação sanguínea , Substância Branca/fisiopatologia , Animais , Infarto Encefálico/metabolismo , Células Endoteliais/metabolismo , Feminino , Proteínas de Fluorescência Verde/genética , Cápsula Interna/irrigação sanguínea , Cápsula Interna/fisiopatologia , Masculino , Camundongos Transgênicos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Substância Branca/metabolismo
6.
Neurochem Int ; 119: 199-206, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29545059

RESUMO

X-ray irradiation (X-irradiation) induces disruption of the blood-brain barrier (BBB). However, the mechanisms underlying the permeability changes are unclear. Therefore, in the present study, we examined the cellular and molecular changes produced by X-irradiation of the brain. Male ICR mice were irradiated locally on their head, posterior to the bregma, except for the eyes, with a single dose of 60 Gy. BBB permeability was assessed using Evans blue dye. We also examined vascular endothelial growth factor (VEGF) expression, microglial morphology, and the expression of the tight junction protein claudin-5 from 0.5 to 7 days after irradiation. An increase in BBB permeability and a decrease in the expression of VEGF protein occurred in a time-dependent manner. In addition, the number of activated microglia (CD68+/Iba-1+ double-positive cells), the amount of tumor necrosis factor-α protein and immunoreactivity of nuclear factor-kappaB increased by irradiation, while the expression of claudin-5 on vascular endothelial cells diminished markedly in the cerebral cortex starting 0.5 days after irradiation. These results suggest that the downregulation of claudin-5 expression mediated by activated microglia may contribute to the BBB disruption induced by X-irradiation.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Claudina-5/metabolismo , Permeabilidade/efeitos dos fármacos , Raios X , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Camundongos Endogâmicos ICR , Microglia/metabolismo , Junções Íntimas/metabolismo
7.
Biochem Biophys Res Commun ; 488(1): 232-238, 2017 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-28499870

RESUMO

We previously reported transplantation of brain microvascular endothelial cells (MVECs) into cerebral white matter infarction model improved the animal's behavioral outcome by increasing the number of oligodendrocyte precursor cells (OPCs). We also revealed extracellular vesicles (EVs) derived from MVECs promoted survival and proliferation of OPCs in vitro. In this study, we investigated the mechanism how EVs derived from MVECs contribute to OPC survival and proliferation. Protein mass spectrometry and enzyme-linked immunosorbent assay revealed fibronectin was abundant on the surface of EVs from MVECs. As fibronectin has been reported to promote OPC survival and proliferation via integrin signaling pathway, we blocked the binding between fibronectin and integrins using RGD sequence mimics. Blocking the binding, however, did not attenuate the survival and proliferation promoting effect of EVs on OPCs. Flow cytometric and imaging analyses revealed fibronectin on EVs mediates their internalization into OPCs by its binding to heparan sulfate proteoglycan on OPCs. OPC survival and proliferation promoted by EVs were attenuated by blocking the internalization of EVs into OPCs. These lines of evidence suggest that fibronectin on EVs mediates their internalization into OPCs, and the cargo of EVs promotes survival and proliferation of OPCs, independent of integrin signaling pathway.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibronectinas/metabolismo , Microvasos/citologia , Oligodendroglia/metabolismo , Células-Tronco/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Integrinas/metabolismo , Masculino , Oligodendroglia/citologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia
8.
FASEB J ; 31(4): 1368-1381, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28007781

RESUMO

We have previously reported that transient receptor potential vanilloid 2 (TRPV2) can be activated by mechanical stimulation, which enhances axonal outgrowth in developing neurons; however, the molecular mechanisms that govern the contribution of TRPV2 activation to axonal outgrowth remain unclear. In the present study, we examined this mechanism by using PC12 cells as a neuronal model. Overexpression of TRPV2 enhanced axonal outgrowth in a mechanical stimulus-dependent manner. Accumulation of TRPV2 at the cell surface was 4-fold greater in the growth cone compared with the soma. In the growth cone, TRPV2 is not static, but dynamically accumulates (within ∼100 ms) to the site of mechanical stimulation. The dynamic and acute clustering of TRPV2 can enhance very weak mechanical stimuli via focal accumulation of TRPV2. Focal application of mechanical stimuli dramatically increased growth cone motility and caused actin reorganization via activation of TRPV2. We also found that TRPV2 physically interacts with actin and that changes in the actin cytoskeleton are required for its activation. Here, we demonstrated for the first time to our knowledge that TRPV2 clustering is induced by mechanical stimulation generated by axonal outgrowth and that TRPV2 activation is triggered by actin rearrangements that result from mechanical stimulation. Moreover, TRPV2 activation enhances growth cone motility and actin accumulation to promote axonal outgrowth. Sugio, S., Nagasawa, M., Kojima, I., Ishizaki, Y., Shibasaki, K. Transient receptor potential vanilloid 2 activation by focal mechanical stimulation requires interaction with the actin cytoskeleton and enhances growth cone motility.


Assuntos
Citoesqueleto de Actina/metabolismo , Cones de Crescimento/metabolismo , Crescimento Neuronal , Canais de Cátion TRPV/metabolismo , Animais , Mecanotransdução Celular , Células PC12 , Ligação Proteica , Ratos , Canais de Cátion TRPV/genética
9.
PLoS One ; 11(7): e0159158, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27403742

RESUMO

We previously examined the effect of brain microvascular endothelial cell (MVEC) transplantation on rat white matter infarction, and found that MVEC transplantation promoted remyelination of demyelinated axons in the infarct region and reduced apoptotic death of oligodendrocyte precursor cells (OPCs). We also found that the conditioned medium (CM) from cultured MVECs inhibited apoptosis of cultured OPCs. In this study, we examined contribution of extracellular vesicles (EVs) contained in the CM to its inhibitory effect on OPC apoptosis. Removal of EVs from the CM by ultracentrifugation reduced its inhibitory effect on OPC apoptosis. To confirm whether EVs derived from MVECs are taken up by cultured OPCs, we labeled EVs with PKH67, a fluorescent dye, and added them to OPC cultures. Many vesicular structures labeled with PKH67 were found within OPCs immediately after their addition. Next we examined the effect of MVEC-derived EVs on OPC behaviors. After 2 days in culture with EVs, there was significantly less pyknotic and more BrdU-positive OPCs when compared to control. We also examined the effect of EVs on motility of OPCs. OPCs migrated longer in the presence of EVs when compared to control. To examine whether these effects on cultured OPCs are shared by EVs from endothelial cells, we prepared EVs from conditioned media of several types of endothelial cells, and tested their effects on cultured OPCs. EVs from all types of endothelial cells we examined reduced apoptosis of OPCs and promoted their motility. Identification of the molecules contained in EVs from endothelial cells may prove helpful for establishment of effective therapies for demyelinating diseases.


Assuntos
Movimento Celular , Sobrevivência Celular , Células Endoteliais/citologia , Vesículas Extracelulares/metabolismo , Oligodendroglia/citologia , Células-Tronco/citologia , Animais , Proliferação de Células , Meios de Cultivo Condicionados , Ratos , Ratos Sprague-Dawley
10.
Neurosci Lett ; 617: 232-5, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-26917099

RESUMO

The Ca(2+)-dependent activator protein for secretion 1 (CAPS1) protein plays a regulatory role in the dense-core vesicle exocytosis pathway. To clarify the functions of this protein in the brain, we searched for novel interaction partners of CAPS1 by mass spectrometry. We identified a specific interaction of CAPS1 with septin family proteins. We also demonstrated that the C-terminal region of the CAPS1 protein binds to part of the deduced GTP-binding domain of septin proteins. It is possible that a tertiary complex of septin, CAPS, and syntaxin contributes to dense-core vesicle trafficking and exocytosis in neurons.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Septinas/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Camundongos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas
11.
Heliyon ; 2(12): e00219, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28054037

RESUMO

AIMS: The prevalence of major depressive disorder (MDD) is higher in women than in men, and this may be due to the decline in estrogen levels that occurs during the menopausal transition. We studied the biological alterations in the medial prefrontal cortex (mPFC), which is a region that is highly implicated in the neurobiology of MDD, and the blood cells (BCs) of ovariectomized (OVX) mice subjected to chronic mild stress (CMS), which represents a mouse model of depression during menopause. MAIN METHODS: The mPFC and the BCs were obtained from the same individuals. Gene expression levels were analyzed by microarray. The data were used for the Ingenuity Pathway Analysis and the Gene Ontology analysis. KEY FINDINGS: The gene expression alterations (GEAs) induced by OVX were mainly associated with ribosomal and mitochondrial functions in both the mPFC and the BCs. Rapamycin-insensitive companion of mTOR (RICTOR) was identified as a possible upstream regulator of the OVX-induced GEAs in both tissues. The CMS-induced GEAs were associated with retinoic acid receptor signaling, inflammatory cytokines and post-synaptic density in the mPFC, but not in the BCs. SIGNIFICANCE: OVX and CMS independently affect biological pathways in the mPFC, which is involved in the development of the depression-like phenotype. Because a subset of the OVX-induced GEAs in the mPFC also occurred in the BCs, the GEAs in the BCs might be a useful probe to predict biological pathways in the corresponding brain tissue under specific conditions such as OVX in females.

12.
J Neurochem ; 135(3): 539-50, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26212499

RESUMO

We previously showed that transplantation of brain microvascular endothelial cells (MVECs) greatly stimulated remyelination in the white matter infarct of the internal capsule (IC) induced by endothelin-1 injection and improved the behavioral outcome. In the present study, we examined the effect of MVEC transplantation on the infarct volume using intermittent magnetic resonance image and on the behavior of oligodendrocyte lineage cells histochemically. Our results in vivo show that MVEC transplantation reduced the infarct volume in IC and apoptotic death of oligodendrocyte precursor cells (OPCs). These results indicate that MVECs have a survival effect on OPCs, and this effect might contribute to the recovery of the white matter infarct. The conditioned-medium from cultured MVECs reduced apoptosis of cultured OPCs, while the conditioned medium from cultured fibroblasts did not show such effect. These results suggest a possibility that transplanted MVECs increased the number of OPCs through the release of humoral factors that prevent their apoptotic death. Identification of such humoral factors may lead to the new therapeutic strategy against ischemic demyelinating diseases.


Assuntos
Isquemia Encefálica/terapia , Doenças Desmielinizantes/terapia , Células Endoteliais/transplante , Microvasos/transplante , Oligodendroglia/fisiologia , Células-Tronco/fisiologia , Animais , Isquemia Encefálica/patologia , Sobrevivência Celular/fisiologia , Doenças Desmielinizantes/patologia , Masculino , Microvasos/citologia , Ratos , Ratos Sprague-Dawley
13.
Am J Physiol Renal Physiol ; 306(12): F1462-76, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24761004

RESUMO

The kidney is one of the major loci for the expression of cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CTH). While CBS-deficient (Cbs(-/-)) mice display homocysteinemia/methioninemia and severe growth retardation, and rarely survive beyond the first 4 wk, CTH-deficient (Cth(-/-)) mice show homocysteinemia/cystathioninemia but develop with no apparent abnormality. This study examined renal amino acid reabsorption in those mice. Although both 2-wk-old Cbs(-/-) and Cth(-/-) mice had normal renal architecture, their serum/urinary amino acid profiles largely differed from wild-type mice. The most striking feature was marked accumulation of Met and cystathionine in serum/urine/kidney samples of Cbs(-/-) and Cth(-/-) mice, respectively. Levels of some neutral amino acids (Val, Leu, Ile, and Tyr) that were not elevated in Cbs(-/-) serum were highly elevated in Cbs(-/-) urine, and urinary excretion of other neutral amino acids (except Met) was much higher than expected from their serum levels, demonstrating neutral aminoaciduria in Cbs(-/-) (not Cth(-/-)) mice. Because the bulk of neutral amino acids is absorbed via a B(0)AT1 transporter and Met has the highest substrate affinity for B(0)AT1 than other neutral amino acids, hypermethioninemia may cause hyperexcretion of neutral amino acids.


Assuntos
Aminoácidos Neutros/metabolismo , Cistationina beta-Sintase/deficiência , Homocistinúria/epidemiologia , Homocistinúria/metabolismo , Aminoacidúrias Renais/epidemiologia , Aminoacidúrias Renais/metabolismo , Animais , Comorbidade , Cistationina/metabolismo , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/deficiência , Cistationina gama-Liase/genética , Modelos Animais de Doenças , Feminino , Hiper-Homocisteinemia/metabolismo , Túbulos Renais Proximais/patologia , Masculino , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
J Biol Chem ; 289(21): 14470-80, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24737318

RESUMO

Astrocytes play active roles in the regulation of synaptic transmission. Neuronal excitation can evoke Ca(2+) transients in astrocytes, and these Ca(2+) transients can modulate neuronal excitability. Although only a subset of astrocytes appears to communicate with neurons, the types of astrocytes that can regulate neuronal excitability are poorly characterized. We found that ∼30% of astrocytes in the brain express transient receptor potential vanilloid 4 (TRPV4), indicating that astrocytic subtypes can be classified on the basis of their expression patterns. When TRPV4(+) astrocytes are activated by ligands such as arachidonic acid, the activation propagates to neighboring astrocytes through gap junctions and by ATP release from the TRPV4(+) astrocytes. After activation, both TRPV4(+) and TRPV4(-) astrocytes release glutamate, which acts as an excitatory gliotransmitter to increase synaptic transmission through type 1 metabotropic glutamate receptor (mGluR). Our results indicate that TRPV4(+) astrocytes constitute a novel subtype of the population and are solely responsible for initiating excitatory gliotransmitter release to enhance synaptic transmission. We propose that TRPV4(+) astrocytes form a core of excitatory glial assembly in the brain and function to efficiently increase neuronal excitation in response to endogenous TRPV4 ligands.


Assuntos
Astrócitos/fisiologia , Ácido Glutâmico/metabolismo , Neurônios/fisiologia , Canais de Cátion TRPV/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Ácido Araquidônico/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores/fisiologia , Expressão Gênica , Células HEK293 , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/citologia , Neuroglia/metabolismo , Neuroglia/fisiologia , Neurônios/metabolismo , Técnicas de Patch-Clamp , Forbóis/farmacologia , Receptores de Glutamato Metabotrópico/metabolismo , Transmissão Sináptica/fisiologia , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/genética
15.
Free Radic Biol Med ; 52(9): 1716-26, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22387178

RESUMO

Physiological roles of the transsulfuration pathway have been recognized by its contribution to the synthesis of cytoprotective cysteine metabolites, such as glutathione, taurine/hypotaurine, and hydrogen sulfide (H(2)S), whereas its roles in protecting against methionine toxicity remained to be clarified. This study aimed at revealing these roles by analyzing high-methionine diet-fed transsulfuration-defective cystathionine γ-lyase-deficient (Cth(-/-)) mice. Wild-type and Cth(-/-) mice were fed a standard diet (1 × Met: 0.44%) or a high-methionine diet (3 × Met or 6 × Met), and hepatic conditions were monitored by serum biochemistry and histology. Metabolome analysis was performed for methionine derivatives using capillary electrophoresis- or liquid chromatography-mass spectrometry and sulfur-detecting gas chromatography. The 6 × Met-fed Cth(-/-) (not 1 × Met-fed Cth(-/-) or 6 × Met-fed wild type) mice displayed acute hepatitis, which was characterized by markedly elevated levels of serum alanine/aspartate aminotransferases and serum/hepatic lipid peroxidation, inflammatory cell infiltration, and hepatocyte ballooning; thereafter, they died of gastrointestinal bleeding due to coagulation factor deficiency. After 1 week on 6 × Met, blood levels of ammonia/homocysteine and hepatic levels of methanethiol/3-methylthiopropionate (a methionine transamination product/methanethiol precursor) became significantly higher in Cth(-/-) mice than in wild-type mice. Although hepatic levels of methionine sulfoxide became higher in 6 × Met-fed wild-type mice and Cth(-/-) mice, those of glutathione, taurine/hypotaurine, and H(2)S became lower and serum levels of homocysteine became much higher in 6 × Met-fed Cth(-/-) mice than in wild-type mice. Thus, transsulfuration plays a critical role in the detoxification of excessive methionine by circumventing aberrant accumulation of its toxic transamination metabolites, including ammonia, methanethiol, and 3-methylthiopropionate, in addition to synthesizing cysteine-derived antioxidants to counteract accumulated pro-oxidants such as methionine sulfoxide and homocysteine.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Cistationina gama-Liase/genética , Dieta , Modelos Animais de Doenças , Hiper-Homocisteinemia/genética , Metionina/administração & dosagem , Aminação , Animais , Células Cultivadas , Cromatografia Gasosa , Cromatografia Líquida , Eletroforese Capilar , Espectrometria de Massas , Metionina/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução
16.
Cerebellum ; 11(1): 181-93, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21732075

RESUMO

Neural stem cells are generally considered to be committed to becoming precursor cells before terminally differentiating into either neurons or glial cells during neural development. Neuronal and oligodendrocyte precursor cells have been identified in several areas in the murine central nervous system. The presence of astrocyte precursor cells (APCs) is not so well understood. The present study provides several lines of evidence that CD44-positive cells are APCs in the early postnatal mouse cerebellum. In developing mouse cerebellum, CD44-positive cells, mostly located in the white matter, were positive for the markers of the astrocyte lineage, but negative for the markers of mature astrocytes. CD44-positive cells were purified from postnatal cerebellum by fluorescence-activated cell sorting and characterized in vitro. In the absence of any signaling molecule, many cells died by apoptosis. The surviving cells gradually expressed glial fibrillary acidic protein, a marker for mature astrocytes, indicating that differentiation into mature astrocytes is the default program for these cells. The cells produced no neurospheres nor neurons nor oligodendrocytes under any condition examined, indicating these cells are not neural stem cells. Leukemia inhibitory factor greatly promoted astrocytic differentiation of CD44-positive cells, whereas bone morphogenetic protein 4 (BMP4) did not. Fibroblast growth factor-2 was a potent mitogen for these cells, but was insufficient for survival. BMP4 inhibited activation of caspase-3 and greatly promoted survival, suggesting a novel role for BMP4 in the control of development of astrocytes in cerebellum. We isolated and characterized only CD44 strongly positive large cells and discarded small and/or CD44 weakly positive cells in this study. Further studies are necessary to characterize these cells to help determine whether CD44 is a selective and specific marker for APCs in the developing mouse cerebellum. In conclusion, we succeeded in preparing APC candidates from developing mouse cerebellum, characterized them in vitro, and found that BMPs are survival factors for these cells.


Assuntos
Astrócitos/metabolismo , Diferenciação Celular/fisiologia , Cerebelo/crescimento & desenvolvimento , Receptores de Hialuronatos/biossíntese , Células-Tronco Neurais/metabolismo , Animais , Animais Recém-Nascidos , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/metabolismo , Astrócitos/citologia , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/fisiologia , Sobrevivência Celular/fisiologia , Cerebelo/citologia , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Cultura Primária de Células/métodos
17.
Neurosci Lett ; 497(2): 116-21, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21536099

RESUMO

Molecules that have crucial functions in both nervous and vascular systems have attracted keen attention recently, and the name "angioneurins" has been proposed. The most striking example of angioneurins is vascular endothelial growth factor A (VEGF), which was originally identified as a key regulator of angiogenesis and has only recently been found to have important functions in the nervous system. In this study, we compared VEGF expression in the vasculature in the brain with that in the aorta and the vasculature in the kidney in mice. In larger vessels containing smooth muscle cells, VEGF was expressed by smooth muscle cells covering the lining of endothelial cells, both in and outside the brain. In cerebral capillaries lacking smooth muscle cells, endothelial cells were closely covered by VEGF-expressing foot processes of astrocytes, whereas capillaries were surrounded by VEGF-expressing processes of podocytes in the renal glomeruli. We also found that cultured cerebral microvessel endothelial cells do not express VEGF, whereas cultured cortical astrocytes do express VEGF.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Comunicação Celular/fisiologia , Células Endoteliais/citologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Animais Recém-Nascidos , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Células Cultivadas , Rim/irrigação sanguínea , Rim/citologia , Camundongos , Camundongos Endogâmicos ICR , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Podócitos/citologia , Podócitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
18.
J Radiat Res ; 52(3): 287-92, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21343678

RESUMO

Adhesion of inflammatory cells to endothelial cells is considered to be involved in the process of radiation-induced damage and fibrosis. Intercellular adhesion molecule-1 (ICAM-1) and transforming growth factor-beta1 (TGF-ß1) are thought to play important roles in this process. In this study, radiation-induced ICAM-1 expression on endothelial cells was investigated with the use of an inhibitor of TGF-ß1 receptor kinase (SB431542) and the effects of X-ray and carbon-ion beam were compared. Cell cultures of human umbilical vein endothelial cells (HUVE cells) were incubated with TGF-ß1 and irradiated with 140 KV X-ray. Next, HUVE cells were irradiated with X-ray and 220 MeV carbon-ion beam with or without SB431542. Immunofluorescence analysis was used to quantify ICAM-1 expression. The expression of ICAM-1 on HUVE cells was significantly increased by the stimulation with TGF-ß1. Expression of ICAM-1 was increased by X-ray and carbon-ion beam irradiation and decreased significantly with SB431542 after both irradiations. The expression of ICAM-1 by 2 Gy of carbon-ion beam irradiation was 6.7 fold higher than that of non-irradiated cells, while 5 Gy of X-ray irradiation increased the expression of ICAM-1 by 2.5 fold. According to ICAM-1 expression, the effect of carbon-ion beam irradiation was about 2.2, 4.4 and 5.0 times greater than that of the same doses of X-ray irradiation (1, 2 and 5 Gy, respectively). The present results suggested that radiation-induced ICAM-1 expression on HUVE cells was, at least partially, regulated by TGF-ß1. Carbon-ion beam induced significantly higher ICAM-1 expression than X-ray.


Assuntos
Células Endoteliais/metabolismo , Células Endoteliais/efeitos da radiação , Molécula 1 de Adesão Intercelular/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Radioisótopos de Carbono , Células Cultivadas , Relação Dose-Resposta à Radiação , Regulação para Baixo/fisiologia , Regulação para Baixo/efeitos da radiação , Íons Pesados , Humanos , Doses de Radiação , Transdução de Sinais/efeitos da radiação , Veias Umbilicais/metabolismo , Veias Umbilicais/efeitos da radiação , Raios X
19.
Hum Mol Genet ; 17(13): 1994-2005, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18364386

RESUMO

Cystathionine beta-synthase-deficient mice (Cbs(-/-)) exhibit several pathophysiological features similar to hyperhomocysteinemic patients, including endothelial dysfunction and hepatic steatosis. Heterozygous mutants (Cbs(+/-)) on the C57BL/6J background are extensively analyzed in laboratories worldwide; however, detailed analyses of Cbs(-/-) have been hampered by the fact that they rarely survive past the weaning age probably due to severe hepatic dysfunction. We backcrossed the mutants with four inbred strains (C57BL/6J(Jcl), BALB/cA, C3H/HeJ and DBA/2J) for seven generations, and compared Cbs(-/-) phenotypes among the different genetic backgrounds. Although Cbs(-/-) on all backgrounds were hyperhomocysteinemic/hypermethioninemic and suffered from lipidosis/hepatic steatosis at 2 weeks of age, >30% of C3H/HeJ-Cbs(-/-) survived over 8 weeks whereas none of DBA/2J-Cbs(-/-) survived beyond 5 weeks. At 2 weeks, serum levels of total homocysteine and triglyceride were lowest in C3H/HeJ-Cbs(-/-). Adult C3H/HeJ-Cbs(-/-) survivors showed hyperhomocysteinemia but escaped hypermethioninemia, lipidosis and hepatic steatosis. They appeared normal in general behavioral tests but showed cerebellar malformation and impaired learning ability in the passive avoidance step-through test, and required sufficient dietary supplementation of cyst(e)ine for survival, demonstrating the essential roles of cystathionine beta-synthase in the central nervous system function and cysteine biosynthesis. Our C3H/HeJ-Cbs(-/-) mice could be useful tools for investigating clinical symptoms such as mental retardation and thromboembolism that are found in homocysteinemic patients.


Assuntos
Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Modelos Animais de Doenças , Hiper-Homocisteinemia/enzimologia , Hiper-Homocisteinemia/genética , Aminoácidos/sangue , Animais , Comportamento Animal , Doenças Cerebelares/enzimologia , Doenças Cerebelares/genética , Doenças Cerebelares/patologia , Doenças Cerebelares/fisiopatologia , Cisteína/metabolismo , Feminino , Humanos , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/fisiopatologia , Estimativa de Kaplan-Meier , Lipídeo A/sangue , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Fosfatidilcolina-Esterol O-Aciltransferase/sangue , Especificidade da Espécie
20.
Neurosci Lett ; 429(2-3): 136-41, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-17997037

RESUMO

To examine lesions caused by seizures in the developing brain, seizures were induced by the intraperitoneal injection of kainate and nicotine into juvenile mice. After a week, whole brain sections were examined using histochemistry and the gene expression profiles in the neocortices and hippocampi were analyzed using a DNA microarray. Propidium iodide and Fluoro-Jade C staining revealed that kainate but not nicotine-induced degeneration of the hippocampal pyramidal neurons. Comparative analyses of 12,488 probe sets on the microarray chip revealed the differential expression of 208 and 1243 probe sets in the neocortices and hippocampi of kainate-injected mice, respectively, as well as that of 535 and 436 probe sets in the neocortices and hippocampi of nicotine-injected mice, respectively, the patterns of change were largely drug-specific and region-specific. Among a variety of kainate-modified genes including those representing neurodegeneration and astrogliosis, we identified an increased gene expression of the lysosomal cysteine protease cathepsin S in the hippocampi of kainate-injected mice. Western blot analysis of the hippocampal homogenates revealed that kainate induced a 3.3-fold increase in cathepsin S expression. Immunohistochemistry using cell type-specific markers showed that cathepsin S was induced in microglia, especially those surrounding degenerating pyramidal neurons, but not in neurons themselves or astroglia, in the hippocampal CA1 region of kainate-injected mice. These results indicate that seizures induced by kainate elicit neurodegeneration, astrogliosis, and microglial activation accompanied by the expression of cathepsin S while those induced by nicotine do not.


Assuntos
Catepsinas/genética , Epilepsia/enzimologia , Regulação Enzimológica da Expressão Gênica/genética , Gliose/enzimologia , Hipocampo/enzimologia , Microglia/enzimologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Catepsinas/metabolismo , Corantes , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Epilepsia/induzido quimicamente , Epilepsia/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Gliose/induzido quimicamente , Gliose/genética , Hipocampo/patologia , Hipocampo/fisiopatologia , Ácido Caínico , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/enzimologia , Degeneração Neural/genética , Neurotoxinas , Nicotina/toxicidade , Agonistas Nicotínicos/toxicidade , Análise de Sequência com Séries de Oligonucleotídeos , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA