16 research outputs found

    Amyloid Beta Peptide Is Released during Thrombosis in the Skin

    No full text
    While it is known that amyloid beta (Aβ) 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 Aβ 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 Aβ40 peptide accumulation in the skin during local experimental photothrombosis in mice. Specifically, Aβ peptide accumulation was concentrated near the dermal blood vessels in thrombotic skin. It was also studied whether the released peptide affects microorganisms. Application of Aβ40 (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 Aβ 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 Aβ release during thrombosis serves as part of a natural defense against infection

    Accumulation of Innate Amyloid Beta Peptide in Glioblastoma Tumors

    No full text
    Immunostaining with specific antibodies has shown that innate amyloid beta (Aβ) is accumulated naturally in glioma tumors and nearby blood vessels in a mouse model of glioma. In immunofluorescence images, Aβ peptide coincides with glioma cells, and enzyme-linked immunosorbent assay (ELISA) have shown that Aβ peptide is enriched in the membrane protein fraction of tumor cells. ELISAs have also confirmed that the Aβ(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 Aβ immunofluorescence coinciding with glioma cells and the nearby vasculature imply that the source of Aβ peptides in glioma can be systemic Aβ from blood vessels, but this question remains unresolved and needs additional studies

    Targeted Delivery of Nanoparticulate Cytochrome C into Glioma Cells Through the Proton-Coupled Folate Transporter

    No full text
    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

    HIV-1 Envelope Protein gp120 Promotes Proliferation and the Activation of Glycolysis in Glioma Cell

    No full text
    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

    Microglia Activate Migration of Glioma Cells through a Pyk2 Intracellular Pathway

    No full text
    <div><p>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.</p></div

    FAK is not involved in microglial stimulation of migration in glioma cells.

    No full text
    <p>Data obtained from migration assays for glioma cells. Cells were treated with 5nM (concentration that effectively blocks FAK) and 16nM (concentration that effectively blocks Pyk2) of PF-562,271. Due to different migration abilities in all presented cell lines the Y-axis scales are adjusted for each cell line in order to demonstrate the absolute numbers of migrating cells. Results are presented as mean 卤 S.D. with significant differences from control in each group (+), from Mock without microglia on the bottom (#), or from Mock with microglia on the bottom (*) (p < 0.05). One-way ANOVA followed by the Tukey鈥檚 multiple comparison test was used to determine significance between groups.</p

    Pyk2 is mostly detected in glioma cells rather than in other cell types in mouse brain.

    No full text
    <p>Immunohistochemistry was performed on C57BL/6 mice brain sections containing the tumor area. GL261 glioma cells were implanted into the brains of C57BL/6 mice and grown for 16 days. Photographs show the tumor and surrounding healthy tissue. The dash line outlines the border of tumor. Anti-GFAP antibody was used to detect glioma cells and astrocytes (red, panel A), anti-Iba 1 antibody was used to detect microglial cells (green, panel B) and Pyk2 detection is presented in blue (panel C). The merged images of anti-GFAP and anti-Pyk2, of anti-Iba1 and anti-Pyk2, and of all antibodies together can be seen in merged image boxes D, E, F correspondingly. Insert panels <b>d</b>, <b>e</b>, and <b>f</b> represent enlarged images of astrocytes, microglia, and invading glioma cells. Solid arrows indicate glioma cells, frame arrows indicate astrocytes, and double headed arrows indicate microglia. Scale bar: 40 渭m.</p

    siRNA knock-down and/or pharmacological blockade of Pyk2 by PF-562,271 eliminates the stimulatory effect of microglia on glioma cell migration.

    No full text
    <p>Data obtained from standard migration assays for control glioma cells and cells transfected with siRNA against Pyk2 with and without additional application of PF-562,271 in the presence and absence of microglia in the lower compartment. Due to different migration abilities in all presented cell lines the Y-axis scales are adjusted for each cell line in order to demonstrate the absolute numbers of migrating cells. Results are presented as mean 卤 S.D. with significant difference from control in each group (+), from Mock without microglia on the bottom (#), or from Mock with microglia on the bottom (*) (p < 0.05). One-way ANOVA followed by the Tukey鈥檚 multiple comparison test was used to determine significance between groups.</p
    corecore