Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 20(10)2019 May 20.
Article in English | MEDLINE | ID: mdl-31137462

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Peptide Fragments/metabolism , Animals , Cell Line, Tumor , Mice , Mice, Inbred C57BL
2.
Biomolecules ; 9(4)2019 04 18.
Article in English | MEDLINE | ID: mdl-31003476

ABSTRACT

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.


Subject(s)
Brain Neoplasms/metabolism , Cytochromes c/chemistry , Glioma/metabolism , Nanoconjugates/chemistry , Proton-Coupled Folate Transporter/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Cytochromes c/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , Mice , Mice, Inbred C57BL , Nanoconjugates/adverse effects
3.
Cancers (Basel) ; 10(9)2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30200472

ABSTRACT

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.

4.
Int J Mol Sci ; 19(6)2018 06 08.
Article in English | MEDLINE | ID: mdl-29890636

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Skin/metabolism , Thrombosis/metabolism , Animals , Astrocytes/metabolism , Cell Membrane/metabolism , Dermis/blood supply , Female , Male , Mice, Inbred C57BL , Saccharomyces cerevisiae/metabolism
5.
Neuroreport ; 28(4): 208-213, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28134630

ABSTRACT

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.


Subject(s)
Connexin 43/metabolism , Gap Junctions/physiology , Neurons/physiology , Polyamines/administration & dosage , Acidosis , Animals , Calcium , Cell Line, Tumor , Gap Junctions/drug effects , Humans , Hydrogen-Ion Concentration , Hypercalcemia , Neurons/metabolism , Rats , Spermine/administration & dosage
6.
J Biol Chem ; 291(14): 7716-26, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26867573

ABSTRACT

TheKCNJ10gene encoding Kir4.1 contains numerous SNPs whose molecular effects remain unknown. We investigated the functional consequences of uncharacterized SNPs (Q212R, L166Q, and G83V) on homomeric (Kir4.1) and heteromeric (Kir4.1-Kir5.1) channel function. We compared these with previously characterized EAST/SeSAME mutants (G77R and A167V) in kidney-derived tsA201 cells and in glial cell-derived C6 glioma cells. The membrane potentials of tsA201 cells expressing G77R and G83V were significantly depolarized as compared with WTKir4.1, whereas cells expressing Q212R, L166Q, and A167V were less affected. Furthermore, macroscopic currents from cells expressing WTKir4.1 and Q212R channels did not differ, whereas currents from cells expressing L166Q, G83V, G77R, and A167V were reduced. Unexpectedly, L166Q current responses were rescued when co-expressed with Kir5.1. In addition, we observed notable differences in channel activity between C6 glioma cells and tsA201 cells expressing L166Q and A167V, suggesting that there are underlying differences between cell lines in terms of Kir4.1 protein synthesis, stability, or expression at the surface. Finally, we determined spermine (SPM) sensitivity of these uncharacterized SNPs and found that Q212R-containing channels displayed reduced block by 1 µmSPM. At 100 µmSPM, the block was equal to or greater than WT, suggesting that the greater driving force of SPM allowed achievement of steady state. In contrast, L166Q-Kir5.1 channels achieved a higher block than WT, suggesting a more stable interaction of SPM in the deep pore cavity. Overall, our data suggest that G83V, L166Q, and Q212R residues play a pivotal role in controlling Kir4.1 channel function.


Subject(s)
Mutation, Missense , Polymorphism, Single Nucleotide , Potassium Channels, Inwardly Rectifying/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Potassium Channels, Inwardly Rectifying/genetics , Rats , Kir5.1 Channel
7.
PLoS One ; 10(6): e0131059, 2015.
Article in English | MEDLINE | ID: mdl-26098895

ABSTRACT

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.


Subject(s)
Brain Neoplasms/physiopathology , Focal Adhesion Kinase 2/physiology , Glioma/physiopathology , Microglia/physiology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/genetics , Gene Knockdown Techniques , Glioma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Rats , Signal Transduction/physiology , Tumor Microenvironment/physiology , Up-Regulation
8.
Neuroreport ; 26(9): 528-32, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26011388

ABSTRACT

Polyamines (PAs), such as spermine and spermidine, modulate the activity of numerous receptors and channels in the central nervous system (CNS) and are stored in glial cells; however, little attention has been paid to their role in the regulation of connexin (Cx)-based gap junction channels. We have previously shown that PAs facilitate diffusion of Lucifer Yellow through astrocytic gap junctions in acute brain slices; therefore, we hypothesized that spermine can regulate Cx43-mediated (as the most abundant Cx in astrocytes) gap junctional communication. We used electrophysiological patch-clamp recording from paired Novikoff cells endogenously expressing Cx43 and HeLaCx43-EGFP transfectants to study pH-dependent modulation of cell-cell coupling in the presence or absence of PAs. Our results showed (i) a higher increase in gap junctional communication at higher concentrations of cytoplasmic spermine, and (ii) that spermine prevented uncoupling of gap junctions at low intracellular pH. Taken together, we conclude that spermine enhances Cx43-mediated gap junctional communication and may preserve neuronal excitability during ischemia and trauma when pH in the brain acidifies. We, therefore, suggest a new role of spermine in the regulation of a Cx43-based network under (patho)physiological conditions.


Subject(s)
Cell Communication/drug effects , Connexin 43/metabolism , Gap Junctions/drug effects , Spermine/pharmacology , Cell Line , Electric Conductivity , HeLa Cells , Humans , Hydrogen-Ion Concentration
9.
J Neurosci Neuroeng ; 3(1): 3-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-25165637

ABSTRACT

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.

10.
Neuroreport ; 23(17): 1021-5, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23076119

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Cell Communication/drug effects , Spermidine/physiology , Spermine/physiology , Animals , Astrocytes/cytology , Astrocytes/drug effects , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/growth & development , Carbenoxolone/pharmacology , Connexin 43/physiology , Female , Fluorescent Dyes/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Giant Cells/drug effects , Giant Cells/metabolism , Intracellular Fluid/metabolism , Isoquinolines/metabolism , Membrane Potentials/drug effects , Rats , Rats, Sprague-Dawley , Spermidine/pharmacology , Spermine/pharmacology
11.
Biotechniques ; 53(5): 305-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23570046

ABSTRACT

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.


Subject(s)
Brain Neoplasms/diagnosis , Glioma/diagnosis , Pyridinium Compounds , Staining and Labeling/methods , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cation Transport Proteins/metabolism , Glioma/metabolism , Glioma/pathology , Humans , In Vitro Techniques , Mice , Microscopy, Confocal , Microscopy, Fluorescence
12.
Epilepsia ; 51(9): 1707-13, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20831751

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels/physiology , Potassium/metabolism , Seizures/genetics , Seizures/metabolism , Amino Acid Substitution/genetics , Amino Acid Substitution/physiology , Animals , Astrocytes/physiology , Barium/pharmacology , Channelopathies/genetics , Channelopathies/metabolism , Channelopathies/physiopathology , Genetic Predisposition to Disease/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiology , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neuroglia/drug effects , Neuroglia/metabolism , Patch-Clamp Techniques , Polymorphism, Single Nucleotide/genetics , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/genetics , Seizures/physiopathology
13.
Open Neurosci J ; 3: 40-47, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19890471

ABSTRACT

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.

14.
Glia ; 56(7): 775-90, 2008 May.
Article in English | MEDLINE | ID: mdl-18293411

ABSTRACT

Although Kir4.1 channels are the major inwardly rectifying channels in glial cells and are widely accepted to support K+- and glutamate-uptake in the nervous system, the properties of Kir4.1 channels during vital changes of K+ and polyamines remain poorly understood. Therefore, the present study examined the voltage-dependence of K+ conductance with varying physiological and pathophysiological external [K+] and intrapipette spermine ([SP]) concentrations in Müller glial cells and in tsA201 cells expressing recombinant Kir4.1 channels. Two different types of [SP] block were characterized: "fast" and "slow." Fast block was steeply voltage-dependent, with only a low sensitivity to spermine and strong dependence on extracellular potassium concentration, [K+]o. Slow block had a strong voltage sensitivity that begins closer to resting membrane potential and was essentially [K+]o-independent, but with a higher spermine- and [K+]i-sensitivity. Using a modified Woodhull model and fitting i/V curves from whole cell recordings, we have calculated free [SP](in) in Müller glial cells as 0.81 +/- 0.24 mM. This is much higher than has been estimated previously in neurons. Biphasic block properties underlie a significantly varying extent of rectification with [K+] and [SP]. While confirming similar properties of glial Kir and recombinant Kir4.1, the results also suggest mechanisms underlying K+ buffering in glial cells: When [K+]o is rapidly increased, as would occur during neuronal excitation, "fast block" would be relieved, promoting potassium influx to glial cells. Increase in [K+]in would then lead to relief of "slow block," further promoting K+-influx.


Subject(s)
Neuroglia/physiology , Neurons/physiology , Potassium Channels, Inwardly Rectifying/physiology , Potassium Channels/physiology , Retina/physiology , Animals , Cells, Cultured , Electrophysiology , Potassium/metabolism , Potassium/pharmacology , Potassium Channels, Inwardly Rectifying/drug effects , Rana pipiens , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Retina/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...