Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 20(10)2019 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-31137462

RESUMO

Immunostaining with specific antibodies has shown that innate amyloid beta (Aß) is accumulated naturally in glioma tumors and nearby blood vessels in a mouse model of glioma. In immunofluorescence images, Aß peptide coincides with glioma cells, and enzyme-linked immunosorbent assay (ELISA) have shown that Aß peptide is enriched in the membrane protein fraction of tumor cells. ELISAs have also confirmed that the Aß(1-40) peptide is enriched in glioma tumor areas relative to healthy brain areas. Thioflavin staining revealed that at least some amyloid is present in glioma tumors in aggregated forms. We may suggest that the presence of aggregated amyloid in glioma tumors together with the presence of Aß immunofluorescence coinciding with glioma cells and the nearby vasculature imply that the source of Aß peptides in glioma can be systemic Aß from blood vessels, but this question remains unresolved and needs additional studies.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Camundongos Endogâmicos C57BL
2.
Int J Mol Sci ; 19(6)2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29890636

RESUMO

While it is known that amyloid beta (Aß) deposits are found in different tissues of both Alzheimer's disease (AD) patients and healthy individuals, there remain questions about the physiological role of these deposits, the origin of the Aß peptide, and the mechanisms of its localization to the tissues. Using immunostaining with specific antibodies, as well as enzyme-linked immunosorbent assay, this study demonstrated Aß40 peptide accumulation in the skin during local experimental photothrombosis in mice. Specifically, Aß peptide accumulation was concentrated near the dermal blood vessels in thrombotic skin. It was also studied whether the released peptide affects microorganisms. Application of Aß40 (4 µM) to the external membrane of yeast cells significantly increased membrane conductance with no visible effect on mouse host cells. The results suggest that Aß release in the skin is related to skin injury and thrombosis, and occurs along with clotting whenever skin is damaged. These results support the proposition that Aß release during thrombosis serves as part of a natural defense against infection.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Pele/metabolismo , Trombose/metabolismo , Animais , Astrócitos/metabolismo , Membrana Celular/metabolismo , Derme/irrigação sanguínea , Feminino , Masculino , Camundongos Endogâmicos C57BL , Saccharomyces cerevisiae/metabolismo
3.
Mol Pharm ; 13(8): 2844-54, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27283751

RESUMO

Proteins often possess highly specific biological activities that make them potential therapeutics, but their physical and chemical instabilities during formulation, storage, and delivery have limited their medical use. Therefore, engineering of nanosized vehicles to stabilize protein therapeutics and to allow for targeted treatment of complex diseases, such as cancer, is of considerable interest. A micelle-like nanoparticle (NP) was designed for both, tumor targeting and stimulus-triggered release of the apoptotic protein cytochrome c (Cyt c). This system is composed of a Cyt c NP stabilized by a folate-receptor targeting amphiphilic copolymer (FA-PEG-PLGA) attached to Cyt c through a redox-sensitive bond. FA-PEG-PLGA-S-S-Cyt c NPs exhibited excellent stability under extracellular physiological conditions, whereas once in the intracellular reducing environment, Cyt c was released from the conjugate. Under the same conditions, the folate-decorated NP reduced folate receptor positive HeLa cell viability to 20%, while the same complex without FA only reduced it to 80%. Confocal microscopy showed that the FA-PEG-PLGA-S-S-Cyt c NPs were internalized by HeLa cells and were capable of endosomal escape. The specificity of the folate receptor-mediated internalization was confirmed by the lack of uptake by two folate receptor deficient cell lines: A549 and NIH-3T3. Finally, the potential as antitumor therapy of our folate-decorated Cyt c-based NPs was confirmed with an in vivo brain tumor model. In conclusion, we were able to create a stable, selective, and smart nanosized Cyt c delivery system.


Assuntos
Citocromos c/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Células A549 , Animais , Apoptose , Citocromos c/química , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Glioma/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Micelas , Células NIH 3T3 , Polímeros/química
4.
J Biol Chem ; 285(46): 36040-8, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20807765

RESUMO

Inwardly rectifying potassium channel Kir4.1 is critical for glial function, control of neuronal excitability, and systemic K(+) homeostasis. Novel mutations in Kir4.1 have been associated with EAST/SeSAME syndrome, characterized by mental retardation, ataxia, seizures, hearing loss, and renal salt waste. Patients are homozygous for R65P, G77R, C140R or T164I; or compound heterozygous for A167V/R297C or R65P/R199Stop, a deletion of the C-terminal half of the protein. We investigated the functional significance of these mutations by radiotracer efflux and inside-out membrane patch clamping in COSm6 cells expressing homomeric Kir4.1 or heteromeric Kir4.1/Kir5.1 channels. All of the mutations compromised channel function, but the underlying mechanisms were different. R65P, T164I, and R297C caused an alkaline shift in pH sensitivity, indicating that these positions are crucial for pH sensing and pore gating. In R297C, this was due to disruption of intersubunit salt bridge Glu(288)-Arg(297). C140R breaks the Cys(108)-Cys(140) disulfide bond essential for protein folding and function. A167V did not affect channel properties but may contribute to decreased surface expression in A167V/R297C. In G77R, introduction of a positive charge within the bilayer may affect channel structure or gating. R199Stop led to a dramatic decrease in surface expression, but channel activity was restored by co-expression with intact subunits, suggesting remarkable tolerance for truncation of the cytoplasmic domain. These results provide an explanation for the molecular defects that underlie the EAST/SeSAME syndrome.


Assuntos
Anormalidades Múltiplas/genética , Ativação do Canal Iônico/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Anormalidades Múltiplas/patologia , Sequência de Aminoácidos , Animais , Ataxia/patologia , Linhagem Celular , Perda Auditiva Neurossensorial/patologia , Humanos , Concentração de Íons de Hidrogênio , Deficiência Intelectual/patologia , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana , Modelos Moleculares , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Multimerização Proteica , Estrutura Terciária de Proteína , Ratos , Convulsões/patologia , Homologia de Sequência de Aminoácidos , Síndrome , Transfecção , Canal Kir5.1
5.
Epilepsia ; 51(9): 1707-13, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20831751

RESUMO

PURPOSE: KCNJ10 encodes subunits of inward rectifying potassium (Kir) channel Kir4.1 found predominantly in glial cells within the brain. Genetic inactivation of these channels in glia impairs extracellular K(+) and glutamate clearance and produces a seizure phenotype. In both mice and humans, polymorphisms and mutations in the KCNJ10 gene have been associated with seizure susceptibility. The purpose of the present study was to determine whether there are differences in Kir channel activity and potassium- and glutamate-buffering capabilities between astrocytes from seizure resistant C57BL/6 (B6) and seizure susceptible DBA/2 (D2) mice that are consistent with an altered K(+) channel activity as a result of genetic polymorphism of KCNJ10. METHODS: Using cultured astrocytes and hippocampal brain slices together with whole-cell patch-clamp, we determined the electrophysiologic properties, particularly K(+) conductances, of B6 and D2 mouse astrocytes. Using a colorimetric assay, we determined glutamate clearance capacity by B6 and D2 astrocytes. RESULTS: Barium-sensitive Kir currents elicited from B6 astrocytes are substantially larger than those elicited from D2 astrocytes. In addition, potassium and glutamate buffering by D2 cortical astrocytes is impaired, relative to buffering by B6 astrocytes. DISCUSSION: In summary, the activity of Kir4.1 channels differs between seizure-susceptible D2 and seizure-resistant B6 mice. Reduced activity of Kir4.1 channels in astrocytes of D2 mice is associated with deficits in potassium and glutamate buffering. These deficits may, in part, explain the relatively low seizure threshold of D2 mice.


Assuntos
Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio/fisiologia , Potássio/metabolismo , Convulsões/genética , Convulsões/metabolismo , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/fisiologia , Animais , Astrócitos/fisiologia , Bário/farmacologia , Canalopatias/genética , Canalopatias/metabolismo , Canalopatias/fisiopatologia , Predisposição Genética para Doença/genética , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Técnicas de Patch-Clamp , Polimorfismo de Nucleotídeo Único/genética , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Convulsões/fisiopatologia
6.
Biomolecules ; 9(4)2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-31003476

RESUMO

In this study, we identified the proton-coupled folate transporter (PCFT) as a route for targeted delivery of drugs to some gliomas. Using the techniques of confocal imaging, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and small interfering (siRNA) knockdown against the PCFT, we demonstrated that Gl261 and A172 glioma cells, but not U87 and primary cultured astrocytes, express the PCFT, which provides selective internalization of folic acid (FA)-conjugated cytochrome c-containing nanoparticles (FA-Cyt c NPs), followed by cell death. The FA-Cyt c NPs (100 µg/mL), had no cytotoxic effects in astrocytes but caused death in glioma cells, according to their level of expression of PCFT. Whole-cell patch clamp recording revealed FA-induced membrane currents in FA-Cyt c NPs-sensitive gliomas, that were reduced by siRNA PCFT knockdown in a similar manner as by application of FA-Cyt c NPs, indicating that the PCFT is a route for internalization of FA-conjugated NPs in these glioma cells. Analysis of human glioblastoma specimens revealed that at least 25% of glioblastomas express elevated level of either PCFT or folate receptor (FOLR1). We conclude that the PCFT provides a mechanism for targeted delivery of drugs to some gliomas as a starting point for the development of efficient methods for treating gliomas with high expression of PCFT and/or FOLR1.


Assuntos
Neoplasias Encefálicas/metabolismo , Citocromos c/química , Glioma/metabolismo , Nanoconjugados/química , Transportador de Folato Acoplado a Próton/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Citocromos c/farmacologia , Ácido Fólico/química , Ácido Fólico/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Nanoconjugados/efeitos adversos
7.
Cancers (Basel) ; 10(9)2018 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-30200472

RESUMO

Patients infected with human immunodeficiency virus (HIV) are more prone to developing cancers, including glioblastomas (GBMs). The median survival for HIV positive GBM patients is significantly shorter than for those who are uninfected, despite the fact that they receive the same treatments. The nature of the GBM⁻HIV association remains poorly understood. In this study, we analyzed the effect of the HIV envelope glycoprotein gp120 on GBM cell proliferation. Specifically, we performed cell cycle, western blot, protein synthesis and metabolomics analysis as well as ATP production and oxygen consumption assays to evaluate proliferation and metabolic pathways in primary human glioma cell line, U87, A172 cells and in the HIVgp120tg/GL261 mouse model. Glioma cells treated with gp120 (100 ng/mL for 7⁻10 days) showed higher proliferation rates and upregulation in the expression of enolase 2, hexokinase and glyceraldehyde-3-phosphate dehydrogenase when compared to untreated cells. Furthermore, we detected an increase in the activity of pyruvate kinase and a higher glycolytic index in gp120 treated cells. Gp120 treated GBM cells also showed heightened lipid and protein synthesis. Overall, we demonstrate that in glioma cells, the HIV envelope glycoprotein promotes proliferation and activation of glycolysis resulting in increased protein and lipid synthesis.

8.
Neuroreport ; 28(4): 208-213, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28134630

RESUMO

Changes in the regulation, formation, and gating of connexin-based gap junction channels occur in various disorders. It has been shown that H and Ca are involved in the regulation of gap junctional communication. Ischemia-induced intracellular acidification and Ca overload lead to closure of gap junctions and inhibit an exchange by ions and small molecules throughout the network of cells in the heart, brain, and other tissues. In this study, we examined the role of the polyamines in the regulation of connexin 43 (Cx43)-based gap junction channels under elevated intracellular concentrations of hydrogen ([H]i) and calcium ([Ca]i) ions. Experiments, conducted in Novikoff and A172 human glioblastoma cells, which endogenously express Cx43, showed that polyamines prevent downregulation of Cx43-mediated gap junctional communication caused by elevated [Ca]i and [H]i, accompanying ischemic and other pathological conditions. siRNA knockdown of Cx43 significantly reduces gap junctional communication, indicating that Cx43 gap junctions are the targets for spermine regulation.


Assuntos
Conexina 43/metabolismo , Junções Comunicantes/fisiologia , Neurônios/fisiologia , Poliaminas/administração & dosagem , Acidose , Animais , Cálcio , Linhagem Celular Tumoral , Junções Comunicantes/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Hipercalcemia , Neurônios/metabolismo , Ratos , Espermina/administração & dosagem
9.
Brain Res Bull ; 128: 98-105, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27908798

RESUMO

INTRODUCTION: Platelets contain beta-amyloid precursor protein (APP) as well as Aß peptide (Aß) that can be released upon activation. During thrombosis, platelets are concentrated in clots and activated. METHODS: We used in vivo fluorescent analysis and electron microscopy in mice to determine to what degree platelets are concentrated in clots. We used immunostaining to visualize Aß after photothrombosis in mouse brains. RESULTS: Both in vivo results and electron microscopy revealed that platelets were 300-500 times more concentrated in clots than in non-clotted blood. After thrombosis in control mice, but not in thrombocytopenic animals, Aß immunofluorescence was present inside blood vessels in the visual cortex and around capillaries in the entorhinal cortex. CONCLUSION: The increased concentration of platelets allows enhanced release of Aß during thrombosis, suggesting an additional source of Aß in the brains of Alzheimer's patients that may arise if frequent micro-thrombosis events occur in their brains.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Plaquetas/metabolismo , Transtornos Cerebrovasculares/metabolismo , Córtex Entorrinal/metabolismo , Trombose/metabolismo , Córtex Visual/metabolismo , Animais , Plaquetas/patologia , Transtornos Cerebrovasculares/patologia , Modelos Animais de Doenças , Córtex Entorrinal/irrigação sanguínea , Córtex Entorrinal/patologia , Feminino , Imuno-Histoquímica , Masculino , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia Eletrônica , Estimulação Luminosa , Contagem de Plaquetas , Trombocitopenia/metabolismo , Trombocitopenia/patologia , Trombose/patologia , Córtex Visual/irrigação sanguínea , Córtex Visual/patologia
10.
PLoS One ; 10(6): e0131059, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098895

RESUMO

Glioblastoma is one of the most aggressive and fatal brain cancers due to the highly invasive nature of glioma cells. Microglia infiltrate most glioma tumors and, therefore, make up an important component of the glioma microenvironment. In the tumor environment, microglia release factors that lead to the degradation of the extracellular matrix and stimulate signaling pathways to promote glioma cell invasion. In the present study, we demonstrated that microglia can promote glioma migration through a mechanism independent of extracellular matrix degradation. Using western blot analysis, we found upregulation of proline rich tyrosine kinase 2 (Pyk2) protein phosphorylated at Tyr579/580 in glioma cells treated with microglia conditioned medium. This upregulation occurred in rodent C6 and GL261 as well as in human glioma cell lines with varying levels of invasiveness (U-87MG, A172, and HS683). siRNA knock-down of Pyk2 protein and pharmacological blockade by the Pyk2/focal-adhesion kinase (FAK) inhibitor PF-562,271 reversed the stimulatory effect of microglia on glioma migration in all cell lines. A lower concentration of PF-562,271 that selectively inhibits FAK, but not Pyk2, did not have any effect on glioma cell migration. Moreover, with the use of the CD11b-HSVTK microglia ablation mouse model we demonstrated that elimination of microglia in the implanted tumors (GL261 glioma cells were used for brain implantation) by the local in-tumor administration of Ganciclovir, significantly reduced the phosphorylation of Pyk2 at Tyr579/580 in implanted tumor cells. Taken together, these data indicate that microglial cells activate glioma cell migration/dispersal through the pro-migratory Pyk2 signaling pathway in glioma cells.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Quinase 2 de Adesão Focal/fisiologia , Glioma/fisiopatologia , Microglia/fisiologia , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Quinase 2 de Adesão Focal/antagonistas & inibidores , Quinase 2 de Adesão Focal/genética , Técnicas de Silenciamento de Genes , Glioma/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Ratos , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Regulação para Cima
11.
PLoS One ; 10(4): e0125195, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25886567

RESUMO

Excitotoxicity due to glutamate receptor over-activation is one of the key mediators of neuronal death after an ischemic insult. Therefore, a major function of astrocytes is to maintain low extracellular levels of glutamate. The ability of astrocytic glutamate transporters to regulate the extracellular glutamate concentration depends upon the hyperpolarized membrane potential of astrocytes conferred by the presence of K+ channels in their membranes. We have previously shown that TREK-2 potassium channels in cultured astrocytes are up-regulated by ischemia and may support glutamate clearance by astrocytes during ischemia. Thus, herein we determine the mechanism leading to this up-regulation and assess the localization of TREK-2 channels in astrocytes after transient middle cerebral artery occlusion. By using a cell surface biotinylation assay we confirmed that functional TREK-2 protein is up-regulated in the astrocytic membrane after ischemic conditions. Using real time RT-PCR, we determined that the levels of TREK-2 mRNA were not increased in response to ischemic conditions. By using Western blot and a variety of protein synthesis inhibitors, we demonstrated that the increase of TREK-2 protein expression requires De novo protein synthesis, while protein degradation pathways do not contribute to TREK-2 up-regulation after ischemic conditions. Immunohistochemical studies revealed TREK-2 localization in astrocytes together with increased expression of the selective glial marker, glial fibrillary acidic protein, in brain 24 hours after transient middle cerebral occlusion. Our data indicate that functional TREK-2 channels are up-regulated in the astrocytic membrane during ischemia through a mechanism requiring De novo protein synthesis. This study provides important information about the mechanisms underlying TREK-2 regulation, which has profound implications in neurological diseases such as ischemia where astrocytes play an important role.


Assuntos
Astrócitos/metabolismo , Ataque Isquêmico Transitório/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Biossíntese de Proteínas , Animais , Astrócitos/patologia , Membrana Celular/metabolismo , Células Cultivadas , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Ataque Isquêmico Transitório/patologia , Canais de Potássio de Domínios Poros em Tandem/genética , Ratos Sprague-Dawley , Regulação para Cima
12.
J Neurosci Neuroeng ; 3(1): 3-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25165637

RESUMO

Organic cation transporters (OCTs) were first found and then isolated from cultured glioma cells. When glioma cells are implanted into brain the fate of OCTs varies with time after implantation and transporter type. Here we report that OCT1, OCT2 and OCT3 immunofluorescence is significantly reduced over time in implanted GL261 glioma cells, during tumor development in the brain. By day 21 after glioma implantation, OCT1, OCT2 and OCT3 immunofluorescence was reduced more than 10-fold in the cytoplasm of glioma cells, while OCT3 immunofluorescence became concentrated in the nucleus. The well-known fluorescent substrate for OCT transporters, 4-(4-(dimethylamino)-styryl)-N-methylpyridinium iodide (ASP+), previously shown to accumulate in glioma-cell cytoplasm in in vivo slices, began to accumulate in the nucleus of these cells, but not in cytoplasm, after 21 days post-implantation. Considering this mislocalization phenomenon, and other literature on similar nuclear mislocalization of different transporters, receptors and channels in glioma cells, we suggest that it is one of the "omens" preceding the motility and aggressivity changes in glioma behavior.

13.
Neuroreport ; 23(17): 1021-5, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23076119

RESUMO

Spermine (SPM) and spermidine, endogenous polyamines with the ability to modulate various ion channels and receptors in the brain, exert neuroprotective, antidepressant, antioxidant, and other effects in vivo such as increasing longevity. These polyamines are preferably accumulated in astrocytes, and we hypothesized that SPM increases glial intercellular communication by interacting with glial gap junctions. The results obtained in situ, using Lucifer yellow propagation in the astrocytic syncitium of 21-25-day-old rat CA1 hippocampal slices, showed reduced coupling when astrocytes were dialyzed with standard intracellular solutions without SPM. However, there was a robust increase in the spreading of Lucifer yellow through gap junctions to neighboring astrocytes when the cells were patched with intracellular solutions containing 1 mM SPM, a physiological concentration in glia. Lucifer yellow propagation was inhibited by gap junction blockers. Our findings show that the glial syncitium propagates SPM through gap junctions and further indicate a new role of polyamines in the regulation of the astroglial network under both normal and pathological conditions.


Assuntos
Astrócitos/metabolismo , Comunicação Celular/efeitos dos fármacos , Espermidina/fisiologia , Espermina/fisiologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/crescimento & desenvolvimento , Carbenoxolona/farmacologia , Conexina 43/fisiologia , Feminino , Corantes Fluorescentes/metabolismo , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Células Gigantes/efeitos dos fármacos , Células Gigantes/metabolismo , Líquido Intracelular/metabolismo , Isoquinolinas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espermidina/farmacologia , Espermina/farmacologia
14.
Biotechniques ; 53(5): 305-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23570046

RESUMO

Here we describe a new method of glioma cell visualization in living brain slices that can be used for evaluation of tumor size or visualization of internal tumor structures. Glial cells, as well as glioma cells of glial origin, express high levels of organic cation transporters. We demonstrate that application of a fluorescent substrate for these transporters 4-(4-(dimethylamino)-styryl)-N-methylpyridinium iodide (ASP+) to the incubation medium leads to quick accumulation of fluorescence in glioma cells during early developmental stages and in astrocytes, but not in neurons. Stained brain slices can be immediately investigated using confocal or fluorescence microscopy. Glioma and glial cells can be discriminated from each other because of their different morphology. The method described has the advantage of staining living tissue and is simple to perform.


Assuntos
Neoplasias Encefálicas/diagnóstico , Glioma/diagnóstico , Compostos de Piridínio , Coloração e Rotulagem/métodos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas de Transporte de Cátions/metabolismo , Glioma/metabolismo , Glioma/patologia , Humanos , Técnicas In Vitro , Camundongos , Microscopia Confocal , Microscopia de Fluorescência
15.
Open Neurosci J ; 3: 40-47, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19890471

RESUMO

The extent of an ischemic insult is less in brain regions enriched in astrocytes suggesting that astrocytes maintain function and buffer glutamate during ischemia. Astrocytes express a wide variety of potassium channels to support their functions including TREK-2 channels which are regulated by polyunsaturated fatty acids, intracellular acidosis and swelling; conditions that pertain to ischemia. The present study investigated the possible involvement of TREK-2 channels in cultured cortical astrocytes during experimental ischemia (anoxia/hypoglycemia) by examining TREK-2 protein levels, channel activity and ability to clear glutamate. We found that TREK-2 protein levels were increased rapidly within 2 hrs of the onset of simulated ischemia. This increase corresponded to an increase in temperature-sensitive TREK-2-like channel conductance and the ability of astrocytes to buffer extracellular glutamate even during ischemia. Together, these data suggest that up-regulation of TREK-2 channels may help rescue astrocyte function and lower extracellular glutamate during ischemia.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA