Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Exp Cell Res ; 408(1): 112857, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34600900

RESUMO

Toxic advanced glycation end products (toxic AGEs) derived from glycolaldehyde (AGE3) have been implicated in the development of diabetic vascular complications such as retinopathy characterised by excessive angiogenesis. Different receptor types, such as receptor for AGEs (RAGE), Toll like receptor-4 and scavenger receptors, are expressed in endothelial cells and contribute to AGE-elicited alteration of cell function. In the present study, we examined the involvement of AGE-related receptors on AGE-induced angiogenesis in endothelial cells. The effects of pharmacological inhibitors or receptor neutralizing antibodies on AGE3-induced tube formation were investigated using the in vitro Matrigel tube formation assay in b.End5 cells (mouse endothelial cells). AGE3-induced signalling pathways and receptor expression changes were analysed by Western blot analysis and flow cytometry, respectively. Both FPS-ZM1, a RAGE inhibitor, and fucoidan, a ligand for scavenger receptors, suppressed AGE3-induced tube formation. Cocktails of neutralizing antibodies against the scavenger receptors CD36, CD163 and LOX-1 prevented AGE3-induced tube formation. AGE3 activated mTOR signalling, resulting in facilitation of tube formation. Activation of the AGE-RAGE pathway also led to the upregulation of scavenger receptors. Taken together, our findings suggest that the scavenger receptors CD36, CD163 and LOX-1 in conjunction with the RAGE receptor work together to mediate toxic AGE-induced facilitation of angiogenesis.


Assuntos
Células Endoteliais/efeitos dos fármacos , Produtos Finais de Glicação Avançada/farmacologia , Neovascularização Patológica/metabolismo , Receptores Depuradores/metabolismo , Animais , Células Endoteliais/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptores Depuradores/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
2.
Biomarkers ; 25(3): 305-313, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32285702

RESUMO

Background: Cerebral small vessel disease (CSVD) is associated with future stroke. Although pathological alteration in small vessels of patients with CSVD can be detected by neuroimaging, diagnosis of CSVD is delayed because it is an asymptomatic disease. The stroke-prone spontaneously hypertensive rat (SHRSP) show similar pathological features to human CSVD and develop stroke-related symptoms with advancing age.Objective: We investigated the time course of haematological parameters in Wistar rats and SHRSP.Material and Methods: Blood cells were analysed using an automated haematological analyser.Results: SHRSP develop stroke-related symptoms including onset of neurological symptoms, decreased body weight and blood brain barrier leakage between 12 and 14 weeks of age. Lymphocyte counts were gradually decreased at 3 weeks before development of stoke-related symptoms and then were further decreased after the development of stroke-related symptoms. The both mean platelet volume and large platelet ratio gradually increased at 3 weeks before the development of stoke-related symptoms. However, although SHRSP showed more microcytic red cells than Wistar rats, the trajectories of change in erythrocyte-related parameters were similar between Wistar rats and SHRSP.Conclusion: Our pilot study suggests that alterations of lymphocyte count and platelet volume predictive indicators for asymptomatic CSVD and symptomatic stroke in SHRSP.


Assuntos
Biomarcadores/sangue , Doenças de Pequenos Vasos Cerebrais/sangue , Hipertensão/sangue , Volume Plaquetário Médio , Acidente Vascular Cerebral/sangue , Animais , Plaquetas/patologia , Doenças de Pequenos Vasos Cerebrais/complicações , Doenças de Pequenos Vasos Cerebrais/diagnóstico , Modelos Animais de Doenças , Humanos , Hipertensão/fisiopatologia , Contagem de Linfócitos , Projetos Piloto , Prognóstico , Ratos Endogâmicos SHR , Ratos Wistar , Sensibilidade e Especificidade , Especificidade da Espécie , Acidente Vascular Cerebral/etiologia , Fatores de Tempo
3.
J Pharmacol Sci ; 138(4): 240-246, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30503674

RESUMO

Cerebral ischemic stress increases cerebral sodium-glucose transporter type 1 (SGLT-1). However, the mechanism by which cerebral ischemia leads to the up-regulation of SGLT-1 remains unclear. In peripheral tissue, the activation of mitogen-activated protein kinases (MAPKs) increases SGLT-1. MAPK pathways [c-Jun N-terminal kinase (JNK), p38 MAPK, and extracellular signal-regulated protein kinase (ERK)] are activated by cerebral ischemic stress. Therefore, we confirmed the involvement of MAPKs in the up-regulation of cerebral SGLT-1 after cerebral ischemia. Male ddY mice were subjected to middle cerebral artery occlusion (MCAO). Protein expression was assessed by western blotting. Mice received an intracerebroventricular (i.c.v.) injection of SP600125 (JNK inhibitor), SB203580 (p38 inhibitor), and PD98059 (MEK inhibitor) immediately after reperfusion. The infarction and behavioral abnormalities were assessed on days 1 and 3 after MCAO. The MAPK inhibitors suppressed the activation of JNK, p38, and ERK 3 h after MCAO. SP600125 and SB203580 administration ameliorated cerebral ischemic neuronal damage, whereas PD98059 administration exacerbated cerebral ischemic neuronal damage. SP600125 and SB203580 significantly suppressed the increase in SGLT-1 12 h after MCAO. PD98059 had no effect on SGLT-1 expression after MCAO. Our results indicate that the activation of JNK and p38 participate in the up-regulation of cerebral SGLT-1 after MCAO.


Assuntos
Infarto da Artéria Cerebral Média/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Neurônios/patologia , Regulação para Cima
4.
ACS Appl Mater Interfaces ; 16(15): 18564-18573, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38567738

RESUMO

Replicating the sense of smell presents an ongoing challenge in the development of biomimetic devices. Olfactory receptors exhibit remarkable discriminatory abilities, including the enantioselective detection of individual odorant molecules. Graphene has emerged as a promising material for biomimetic electronic devices due to its unique electrical properties and exceptional sensitivity. However, the efficient detection of nonpolar odor molecules using transistor-based graphene sensors in a gas phase in environmental conditions remains challenging due to high sensitivity to water vapor. This limitation has impeded the practical development of gas-phase graphene odor sensors capable of selective detection, particularly in humid environments. In this study, we address this challenge by introducing peptide-functionalized graphene sensors that effectively mitigate undesired responses to changes in humidity. Additionally, we demonstrate the significant role of humidity in facilitating the selective detection of odorant molecules by the peptides. These peptides, designed to mimic a fruit fly olfactory receptor, spontaneously assemble into a monomolecular layer on graphene, enabling precise and specific odorant detection. The developed sensors exhibit notable enantioselectivity, achieving a remarkable 35-fold signal contrast between d- and l-limonene. Furthermore, these sensors display distinct responses to various other biogenic volatile organic compounds, demonstrating their versatility as robust tools for odor detection. By acting as both a bioprobe and an electrical signal amplifier, the peptide layer represents a novel and effective strategy to achieve selective odorant detection under normal atmospheric conditions using graphene sensors. This study offers valuable insights into the development of practical odor-sensing technologies with potential applications in diverse fields.


Assuntos
Técnicas Biossensoriais , Grafite , Receptores Odorantes , Odorantes , Grafite/química , Gases , Estereoisomerismo , Receptores Odorantes/química , Peptídeos
5.
J Pharmacol Exp Ther ; 344(1): 276-85, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23117790

RESUMO

Orexin-A (a glucose-sensing neuropeptide in the hypothalamus) and brain-derived neurotrophic factor (BDNF; a member of the neurotrophin family) play roles in many physiologic functions, including regulation of glucose metabolism. We previously showed that the development of postischemic glucose intolerance is one of the triggers of ischemic neuronal damage. The aim of this study was to determine whether there was an interaction between orexin-A and BDNF functions in the hypothalamus after cerebral ischemic stress. Male ddY mice were subjected to 2 hours of middle cerebral artery occlusion (MCAO). Neuronal damage was estimated by histologic and behavioral analyses. Expression of protein levels was analyzed by Western blot. Small interfering RNA directed BDNF, orexin-A, and SB334867 [N-(2-methyl-6-benzoxazolyl)-N'-1,5-naphthyridin-4-yl urea; a specific orexin-1 receptor antagonist] were administered directly into the hypothalamus. The level of hypothalamic orexin-A, detected by immunohistochemistry, was decreased on day 1 after MCAO. Intrahypothalamic administration of orexin-A (1 or 5 pmol/mouse) significantly and dose-dependently suppressed the development of postischemic glucose intolerance on day 1 and development of neuronal damage on day 3. The MCAO-induced decrease in insulin receptor levels in the liver and skeletal muscle on day 1 was recovered to control levels by orexin-A, and this effect of orexin-A was reversed by the administration of SB334867 as well as by hypothalamic BDNF knockdown. These results suggest that suppression of postischemic glucose intolerance by orexin-A assists in the prevention of cerebral ischemic neuronal damage. In addition, hypothalamic BDNF may play an important role in this effect of orexin-A.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Intolerância à Glucose/tratamento farmacológico , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neurônios/patologia , Neuropeptídeos/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Glicemia/metabolismo , Western Blotting , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Fator Neurotrófico Derivado do Encéfalo/genética , Infarto Cerebral/patologia , Intolerância à Glucose/etiologia , Glucosefosfato Desidrogenase/metabolismo , Hipotálamo/efeitos dos fármacos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Masculino , Memória/efeitos dos fármacos , Camundongos , Microinjeções , Exame Neurológico , Neuropeptídeos/administração & dosagem , Orexinas , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Receptor de Insulina/biossíntese
6.
Diab Vasc Dis Res ; 17(1): 1479164119896975, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32000529

RESUMO

Advanced glycation end-products, especially toxic advanced glycation end-products derived from glyceraldehyde (advanced glycation end-product-2) and glycolaldehyde (advanced glycation end-product-3), are biologically reactive compounds associated with diabetic complications. We previously demonstrated that toxic advanced glycation end-products were internalised into macrophage-like RAW264.7 cells through scavenger receptor-1 class A (CD204). Toxic advanced glycation end-product uptake was inhibited by fucoidan, a sulphated polysaccharide and antagonistic ligand for scavenger receptors, suggesting that sulphated polysaccharides are emerging candidates for treatment of advanced glycation end-product-related diseases. In this study, we compared the effects of six types of sulphated and non-sulphated polysaccharides on toxic advanced glycation end-product uptake in RAW264.7 cells. Fucoidan, carrageenan and dextran sulphate attenuated toxic advanced glycation end-product uptake. Fucoidan and carrageenan inhibited advanced glycation end-product-2-induced upregulation of SR-A, while advanced glycation end-product-3-induced upregulation of scavenger receptor-1 class A was only suppressed by fucoidan. Dextran sulphate did not affect scavenger receptor-1 class A levels in toxic advanced glycation end-product-treated cells. Chondroitin sulphate, heparin and hyaluronic acid failed to attenuate toxic advanced glycation end-product uptake. Heparin and hyaluronic acid had no effect on scavenger receptor-1 class A levels, while chondroitin sulphate inhibited advanced glycation end-product-3-induced upregulation of scavenger receptor-1 class A. Taken together, fucoidan and carrageenan, but not the other sulphated polysaccharides examined, had inhibitory activities on toxic advanced glycation end-product uptake and toxic advanced glycation end-product-induced upregulation of scavenger receptor-1 class A, possibly because of structural differences among sulphated polysaccharides.


Assuntos
Carragenina/farmacologia , Produtos Finais de Glicação Avançada/metabolismo , Macrófagos/efeitos dos fármacos , Polissacarídeos/farmacologia , Receptores Depuradores Classe A/antagonistas & inibidores , Animais , Transporte Biológico , Sulfatos de Condroitina/farmacologia , Sulfato de Dextrana/farmacologia , Heparina/farmacologia , Ácido Hialurônico/farmacologia , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Receptores Depuradores Classe A/metabolismo
7.
Yakugaku Zasshi ; 138(7): 955-962, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-29962475

RESUMO

 Cerebral ischemic stress often induces a hyperglycemic condition. This postischemic hyperglycemia exacerbates the development of cerebral ischemic neuronal damage, although the mechanism of this exacerbation remains to be clarified. We previously discovered that the cerebral sodium-glucose transporter (SGLT) was closely involved in the development of cerebral ischemic neuronal damage. SGLT is a member of the glucose transporter family and moves glucose together with sodium ions. SGLT-1, -3, -4, and -6 are distributed in the brain. We conducted further experiments to elucidate the detailed mechanism of the exacerbation of cerebral ischemia by cerebral SGLT. The results clarified: 1) the relationship between cerebral SGLT and postischemic hyperglycemia; 2) the involvement of cerebral SGLT-1 (a cerebral SGLT isoform) in cerebral ischemic neuronal damage; and 3) the effects of sodium influx through cerebral SGLT on the development of cerebral ischemic neuronal damage. This paper presents our data on the involvement of cerebral SGLT in the exacerbation of cerebral ischemic neuronal damage.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/genética , Terapia de Alvo Molecular , Proteínas de Transporte de Sódio-Glucose , Animais , Encéfalo/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Glucose/metabolismo , Humanos , Hiperglicemia/etiologia , Camundongos , Neurônios/patologia , Sódio/metabolismo , Proteínas de Transporte de Sódio-Glucose/metabolismo , Proteínas de Transporte de Sódio-Glucose/fisiologia
8.
Eur J Pharmacol ; 822: 25-31, 2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29329760

RESUMO

Glucose is the primary energy fuel of life. A glucose transporter, the sodium-glucose transporter (SGLT), is receiving attention as a novel therapeutic target in disease. This review summarizes the physiological role of SGLT in cerebral ischemia, cancer, cardiac disease, and intestinal ischemia, which has encouraged analysis of SGLT function. In cerebral ischemia and cardiomyopathy, SGLT-1 is involved in worsening of the injury. In addition, SGLT-1 promotes the development of cancer. On the other hand, SGLT-1 has a protective effect against cardiac and intestinal ischemia. Interestingly, SGLT-1 expression levels are increased in some diseased tissue, such as in cerebral ischemia and cancer. This suggests that SGLT-1 may have an important role in many diseases. This review discusses the potential of SGLT as a target for novel therapeutic agents.


Assuntos
Terapia de Alvo Molecular/métodos , Proteínas de Transporte de Sódio-Glucose/metabolismo , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia
9.
Sci Rep ; 8(1): 5901, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29651042

RESUMO

Advanced glycation end-products (AGEs), which comprise non-enzymatically glycosylated proteins, lipids, and nucleic acid amino groups, play an important role in several diseases and aging processes including angiopathy, renal failure, diabetic complications, and neurodegenerative diseases. Among AGE-associated phenotypes, toxic AGEs, glyceraldehyde-derived AGE-2, and glycolaldehyde-derived AGE-3 are involved in the pathogenesis of diabetic complications. In addition, macrophages are reported to remove extracellular AGEs from tissues via scavenger receptors, leading to the progression of atherosclerosis. In the present study, we found that AGE-2 and AGE-3 enhanced their own endocytic uptake by RAW264.7 mouse macrophage-like cells in a concentration-dependent manner. Furthermore, we demonstrated, for the first time, the morphology of phagocytic macrophages and the endocytosis of AGE particles. The toxic AGEs induced the expression of a scavenger receptor, CD204/scavenger receptors-1 class A (SR-A). Notably, an antibody against CD204 significantly prevented toxic AGE uptake. Moreover, an SR-A antagonistic ligand, fucoidan, also attenuated the AGE-2- and AGE-3-evoked uptake in a concentration-dependent manner. These results indicated that SR-A stimulation, at least in part, plays a role in AGE uptake.


Assuntos
Acetaldeído/análogos & derivados , Produtos Finais de Glicação Avançada/genética , Gliceraldeído/metabolismo , Processamento de Proteína Pós-Traducional , Receptores Depuradores Classe A/genética , Acetaldeído/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Endocitose/efeitos dos fármacos , Regulação da Expressão Gênica , Produtos Finais de Glicação Avançada/agonistas , Produtos Finais de Glicação Avançada/imunologia , Camundongos , Fagocitose/efeitos dos fármacos , Polissacarídeos/farmacologia , Células RAW 264.7 , Receptores Depuradores Classe A/antagonistas & inibidores , Receptores Depuradores Classe A/imunologia
10.
Front Immunol ; 9: 334, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29559970

RESUMO

M2 macrophage (Mφ) promotes pathologic angiogenesis through a release of pro-angiogenic mediators or the direct cell-cell interaction with endothelium in the micromilieu of several chronic inflammatory diseases, including rheumatoid arthritis and cancer, where interleukin (IL)-18 also contributes to excessive angiogenesis. However, the detailed mechanism remains unclear. The aim of this study is to investigate the mechanism by which M2 Mφs in the micromilieu containing IL-18 induce excessive angiogenesis in the in vitro experimental model using mouse Mφ-like cell line, RAW264.7 cells, and mouse endothelial cell line, b.End5 cells. We discovered that IL-18 acts synergistically with IL-10 to amplify the production of Mφ-derived mediators like osteopontin (OPN) and thrombin, yielding thrombin-cleaved form of OPN generation, which acts through integrins α4/α9, thereby augmenting M2 polarization of Mφ with characteristics of increasing surface CD163 expression in association with morphological alteration. Furthermore, the results of visualizing temporal behavior and morphological alteration of Mφs during angiogenesis demonstrated that M2-like Mφs induced excessive angiogenesis through the direct cell-cell interaction with endothelial cells, possibly mediated by CD163.


Assuntos
Comunicação Celular/imunologia , Polaridade Celular/imunologia , Células Endoteliais/imunologia , Interleucina-18/imunologia , Macrófagos/imunologia , Neovascularização Patológica/imunologia , Animais , Linhagem Celular Tumoral , Células Endoteliais/patologia , Interleucina-10/imunologia , Macrófagos/patologia , Camundongos , Neovascularização Patológica/patologia , Osteopontina/imunologia , Células RAW 264.7 , Trombina/imunologia
11.
Brain Res ; 1661: 100-110, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28223018

RESUMO

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the authors following an investigation into data manipulation (Fig.3A-D and Fig.4A-F) by an Investigation Committee at Kobe Gakuin University. Namely: Fig.3A-D and Fig.4A-F ­ numerical disagreement (numbers removed) was found in some parts between the raw data and the article data, hence the significant difference illustrated in the published article was not obtained.


Assuntos
Orexinas/metabolismo , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia , Animais , Isquemia Encefálica/complicações , Intolerância à Glucose/complicações , Doenças Hipotalâmicas , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Infarto da Artéria Cerebral Média/complicações , Inflamação , Insulina/farmacologia , Interleucina-1beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Fígado/metabolismo , Masculino , Bulbo/metabolismo , Bulbo/fisiologia , Memória/efeitos dos fármacos , Camundongos , Neurônios/metabolismo , Orexinas/efeitos dos fármacos , Receptor de Insulina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Nervo Vago/metabolismo
12.
Eur J Pharmacol ; 799: 103-110, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28174043

RESUMO

We recently reported that cerebral sodium-glucose transporter type 1 (SGLT-1) plays a role in exacerbation of cerebral ischemia. However, the mechanism by which cerebral SGLT-1 acts remains unclear. Here we demonstrated that sodium influx through cerebral SGLT-1 exacerbates cerebral ischemic neuronal damage. SGLT-specific sodium ion influx was induced using α-methyl-D-glucopyranoside (α-MG). Intracellular sodium concentrations in primary cortical neurons were estimated using sodium-binding benzofuran isophthalate fluorescence. SGLT-1 knockdown in primary cortical neurons and mice was achieved using SGLT-1 siRNA. The survival rates of primary cultured cortical neurons were assessed using biochemical assays 1 day after treatment. Middle cerebral artery occlusion (MCAO) was used to generate a focal cerebral ischemic model in SGLT-1 knockdown mice. The change in fasting blood glucose levels, infarction development, and behavioral abnormalities were assessed 1 day after MCAO. Treatment with 200mM α-MG induced a continuous increase in the intracellular sodium concentration, and this increase was normalized after α-MG removal. Neuronal SGLT-1 knockdown had no effect on 100µM H2O2-induced neuronal cell death; however, the knockdown prevented the neuronal cell death induced by 17.5mM glucose and the co-treatment of 100µM H2O2/8.75mM glucose. Neuronal SGLT-1 knockdown also suppressed the cell death induced by α-MG alone and the co-treatment of 100µM H2O2/0.01mM α-MG. Our in vivo results showed that the exacerbation of cerebral ischemic neuronal damage induced by the intracerebroventricular administration of 5.0µg α-MG/mouse was ameliorated in cerebral SGLT-1 knockdown mice. Thus, sodium influx through cerebral SGLT-1 may exacerbate cerebral ischemia-induced neuronal damage.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Neurônios/patologia , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Hiperglicemia/complicações , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Masculino , Metilglucosídeos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , RNA Interferente Pequeno/genética , Proteínas de Transporte de Sódio-Glucose/metabolismo , Transportador 1 de Glucose-Sódio/deficiência , Transportador 1 de Glucose-Sódio/genética
13.
J Pharm Pharmacol ; 68(7): 922-31, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27139580

RESUMO

OBJECTIVES: We recently demonstrated that the cerebral sodium-glucose transporter (SGLT) is involved in postischaemic hyperglycaemia-induced exacerbation of cerebral ischaemia. However, the associated SGLT-mediated mechanisms remain unclear. Thus, we examined the involvement of cerebral SGLT-induced excessive sodium ion influx in the development of cerebral ischaemic neuronal damage. METHODS: [Na+]i was estimated according to sodium-binding benzofuran isophthalate fluorescence. In the in vitro study, primary cortical neurons were prepared from fetuses of ddY mice. Primary cortical neurons were cultured for 5 days before each treatment with reagents, and these survival rates were assessed using biochemical assays. In in vivo study, a mouse model of focal ischaemia was generated using middle cerebral artery occlusion (MCAO). KEY FINDINGS: In these experiments, treatment with high concentrations of glucose induced increment in [Na+]i, and this phenomenon was suppressed by the SGLT-specific inhibitor phlorizin. SGLT-specific sodium ion influx was induced using a-methyl-D-glucopyranoside (a-MG) treatments, which led to significant concentration-dependent declines in neuronal survival rates and exacerbated hydrogen peroxide-induced neuronal cell death. Moreover, phlorizin ameliorated these effects. Finally, intracerebroventricular administration of a-MG exacerbated the development of neuronal damage induced by MCAO, and these effects were ameliorated by the administration of phlorizin. CONCLUSIONS: Hence, excessive influx of sodium ions into neuronal cells through cerebral SGLT may exacerbate the development of cerebral ischaemic neuronal damage.


Assuntos
Córtex Cerebral/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Animais , Glicemia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glucose/antagonistas & inibidores , Glucose/farmacologia , Peróxido de Hidrogênio/efeitos adversos , Peróxido de Hidrogênio/antagonistas & inibidores , Infarto da Artéria Cerebral Média/patologia , Infusões Intraventriculares , Metilglucosídeos/administração & dosagem , Metilglucosídeos/antagonistas & inibidores , Metilglucosídeos/farmacologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Florizina/administração & dosagem , Florizina/farmacologia , Cultura Primária de Células
15.
Neurosci Lett ; 604: 134-9, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26254165

RESUMO

Post-ischemic hyperglycemia exacerbates the development of cerebral ischemia. To elucidate this exacerbation mechanism, we focused on sodium-glucose transporter (SGLT) as a mediator that lead hyperglycemia to cerebral ischemia. SGLT transport glucose into the cell, together with sodium ion, using the sodium concentration gradient. We have previously reported that suppression of cerebral SGLT ameliorates cerebral ischemic neuronal damage. However, detail relationship cerebral between SGLT and post-ischemic hyperglycemia remain incompletely defined. Therefore, we examined the involvement of cerebral SGLT on cerebral ischemic neuronal damage with or without hyperglycemic condition. Cell survival rate of primary cultured neurons was assessed by biochemical assay. A mouse model of focal ischemia was generated using a middle cerebral artery occlusion (MCAO). Neuronal damage was assessed with histological and behavioral analyses. Concomitant hydrogen peroxide/glucose treatment exacerbated hydrogen peroxide alone-induced cell death. Although a SGLT family-specific inhibitor, phlorizin had no effect on developed hydrogen peroxide alone-induced cell death, it suppressed cell death induced by concomitant hydrogen peroxide/glucose treatment. α-MG induced a concentration-dependent and significant decrease in neuronal survival. PHZ administered on immediately after reperfusion had no effect, but PHZ given at 6h after reperfusion had an effect. Our in vitro study indicates that SGLT is not involved in neuronal cell death in non-hyperglycemic condition. We have already reported that post-ischemic hyperglycemia begins to develop at 6h after MCAO. Therefore, current our in vivo study show post-ischemic hyperglycemic condition may be necessary for the SGLT-mediated exacerbation of cerebral ischemic neuronal damage.


Assuntos
Córtex Cerebral/metabolismo , Hiperglicemia/metabolismo , Ataque Isquêmico Transitório/metabolismo , Proteínas de Transporte de Sódio-Glucose/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Córtex Cerebral/patologia , Peróxido de Hidrogênio/farmacologia , Hiperglicemia/etiologia , Hiperglicemia/patologia , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Neurônios/patologia , Florizina/farmacologia , Proteínas de Transporte de Sódio-Glucose/antagonistas & inibidores , Fatores de Tempo
16.
PLoS One ; 9(4): e95433, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24759941

RESUMO

Orexin-A (a neuropeptide in the hypothalamus) plays an important role in many physiological functions, including the regulation of glucose metabolism. We have previously found that the development of post-ischemic glucose intolerance is one of the triggers of ischemic neuronal damage, which is suppressed by hypothalamic orexin-A. Other reports have shown that the communication system between brain and peripheral tissues through the autonomic nervous system (sympathetic, parasympathetic and vagus nerve) is important for maintaining glucose and energy metabolism. The aim of this study was to determine the involvement of the hepatic vagus nerve on hypothalamic orexin-A-mediated suppression of post-ischemic glucose intolerance development and ischemic neuronal damage. Male ddY mice were subjected to middle cerebral artery occlusion (MCAO) for 2 h. Intrahypothalamic orexin-A (5 pmol/mouse) administration significantly suppressed the development of post-ischemic glucose intolerance and neuronal damage on day 1 and 3, respectively after MCAO. MCAO-induced decrease of hepatic insulin receptors and increase of hepatic gluconeogenic enzymes on day 1 after was reversed to control levels by orexin-A. This effect was reversed by intramedullary administration of the orexin-1 receptor antagonist, SB334867, or hepatic vagotomy. In the medulla oblongata, orexin-A induced the co-localization of cholin acetyltransferase (cholinergic neuronal marker used for the vagus nerve) with orexin-1 receptor and c-Fos (activated neural cells marker). These results suggest that the hepatic branch vagus nerve projecting from the medulla oblongata plays an important role in the recovery of post-ischemic glucose intolerance and mediates a neuroprotective effect by hypothalamic orexin-A.


Assuntos
Intolerância à Glucose/metabolismo , Hipotálamo/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Fígado/inervação , Neuropeptídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Nervo Vago/fisiologia , Animais , Benzoxazóis/farmacologia , Colina O-Acetiltransferase/metabolismo , Hipotálamo/efeitos dos fármacos , Masculino , Camundongos , Naftiridinas , Receptores de Orexina/metabolismo , Orexinas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ureia/análogos & derivados , Ureia/farmacologia , Nervo Vago/efeitos dos fármacos
17.
Brain Res ; 1541: 61-8, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24134951

RESUMO

Diabetes mellitus and impaired glucose metabolism are the most important risk factors for stroke. We recently demonstrated that cerebral ischemic stress causes hyperglycemia (i.e., post-ischemic hyperglycemia) and may worsen ischemic neuronal damage in a mouse model of focal ischemia. However, the detailed mechanisms are still unknown. The sodium-glucose transporter (SGLT) generates inward currents in the process of transporting glucose into cells, resulting in depolarization and increased excitability, which is well known to be caused by cerebral ischemia. Hence, we focused on the role of SGLT on the development of neuronal damage using a global ischemic model. Male ddY mice were subjected to 30min of bilateral carotid artery occlusion (BCAO). The neuronal damage was estimated by histological analysis using HE staining on day 3 after BCAO. Intraperitoneal (i.p.) administration of phlorizin (a specific and competitive inhibitor of SGLT, 200mg/kg immediately after reperfusion) suppressed the development of post-ischemic hyperglycemia on day 1 after BCAO. In contrast, intracerebroventricular (i.c.v.) administration of phlorizin (40µg/mouse immediately and 6h after reperfusion) had no effect on day 1 after BCAO. Interestingly, the development of ischemic neuronal damage was significantly suppressed by i.p. and i.c.v. administration of phlorizin on day 3 after BCAO. In addition, BCAO-induced spasticity was significantly suppressed by PHZ (40µg/mouse, i.c.v.) from using gait analysis. Our results indicated that cerebral SGLT was involved in the development of ischemic neuronal damage in global ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Fármacos Neuroprotetores/farmacologia , Florizina/farmacologia , Proteínas de Transporte de Sódio-Glucose/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Proteínas de Transporte de Sódio-Glucose/efeitos dos fármacos
18.
Brain Res ; 1489: 113-20, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23078759

RESUMO

Post-ischemic hyperglycemia may be one of the triggers of ischemic neuronal damage. However, the detailed mechanisms of this injury process are still unknown. Here, we focused on the involvement of the sodium-glucose transporter (SGLT), which transports glucose together with Na(+) ions, and generates inward currents while transporting glucose into cells, resulting in depolarization and increased excitability. The aim of this study was to determine the involvement of the SGLT in the development of cerebral ischemic stress-induced neuronal damage. Male ddY mice were subjected to 2h of middle cerebral artery occlusion (MCAO). Fasting blood glucose (FBG) was measured using the glucose pilot. Neuronal damage was estimated by histological and behavioral analyses. Phlorizin and glucose were administered by intraperitoneal (i.p.) or intracerebroventricular (i.c.v.) injection. Administration of phlorizin (40, 120 or 200mg/kg, i.p.) significantly and dose-dependently suppressed the elevation of FBG and ischemic neuronal damage. In contrast, phlorizin (10 or 40µg/mouse, i.c.v.) significantly and dose-dependently suppressed ischemic neuronal damage without reducing the elevation of FBG. Moreover, the development of neuronal damage was significantly and dose-dependently exacerbated following i.c.v. administration of glucose (10% or 25% (w/v)), and its exacerbation was suppressed by i.c.v. administration of phlorizin (40µg/mouse). These results suggest that cerebral SGLT is activated by post-ischemic hyperglycemia and may be involved in the exacerbation of ischemic neuronal damage.


Assuntos
Isquemia Encefálica/metabolismo , Hiperglicemia/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Animais não Endogâmicos , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glucose/farmacologia , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Hiperglicemia/patologia , Infarto da Artéria Cerebral Média/patologia , Injeções Intraperitoneais , Injeções Intraventriculares , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Florizina/farmacologia , Estresse Fisiológico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA