31 research outputs found

    Preliminary studies on the behavioural effects of the methanol extract of Leonotis nepetifolia Linn stem in mice

    Get PDF
    Background: Leonotis nepetifolia Linn (Lamiaceae) is used in traditional medicine for its calming (tranquilizing) effects. The aim of this study was to determine whether there is any scientific justification for this use. To achieve this purpose, we investigated the behavioural effects of the methanol extract of Leonotis nepetifolia stem (37.5, 75 and 150 mg/kg) in mice.Methods: Acute toxicity studies were carried out on the methanol stem extract of Leonotis nepetifolia to determine the LD50. The behavioural tests employed were diazepam-induced sleep onset and duration, hole board assay for exploratory activity, mouse beam walk assay for motor coordination, and the staircase test for the detection of anxiolytic compounds. Preliminary phytochemical screening was also carried out on the extract.Results: The intraperitoneal LD50 value was found to be 3.8 g/kg. The results showed that the extract significantly prolonged the duration of diazepam-induced sleep at the highest dose (150 mg/kg). There was no observable effect on exploratory activity and motor coordination at the doses tested (37.5, 75 and 150 mg/kg). The extract, however, at 150 mg/kg elicited a significant decrease in the number of rearings in the staircase test, an effect also observed in the group of mice injected with an anxiolytic dose of diazepam. The preliminary phytochemical  screening revealed the presence of alkaloids, saponins, glycosides and   triterpenoids.Conclusion: The results obtained suggest that the crude methanol extract of Leonotis nepetifolia stem possesses some biologically active constituents with potential anxiolytic activity and thus may justify its traditional use as a tranquilizer.Keywords: behavioural; exploratory; Leonotis nepetifolia; motor coordination; anxiolyti

    Regulation of smooth muscle α-actin expression and hypertrophy in cultured mesangial cells

    Get PDF
    Regulation of smooth muscle α-actin expression and hypertrophy in cultured mesangial cells.BackgroundMesangial cells during embryonic development and glomerular disease express smooth muscle α-actin (α-SMA). We were therefore surprised when cultured mesangial cells deprived of serum markedly increased expression of α-SMA. Serum-deprived mesangial cells appeared larger than serum-fed mesangial cells. We hypothesized that α-SMA expression may be more reflective of mesangial cell hypertrophy than hyperplasia.MethodsHuman mesangial cells were cultured in medium alone or with fetal bovine serum, thrombin, platelet-derived growth factor-BB (PDGF-BB) and/or transforming growth factor-β1 (TGF-β1). α-SMA expression was examined by immunofluorescence, Western blot, and Northern blot analysis. Cell size was analyzed by forward light scatter flow cytometry.Resultsα-SMA mRNA was at least tenfold more abundant after three to five days in human mesangial cells plated without serum, but β-actin mRNA was unchanged. Serum-deprived cells contained 5.3-fold more α-SMA after three days and 56-fold more after five days by Western blot. Serum deprivation also increased α-SMA in rat and mouse mesangial cells. The effects of serum deprivation on α-SMA expression were reversible. Mesangial cell mitogens, thrombin or PDGF-BB, decreased α-SMA, but TGF-β1 increased α-SMA expression and slowed mesangial cell proliferation in serum-plus medium. Flow cytometry showed that serum deprivation or TGF-β1 treatment caused mesangial cell hypertrophy. PDGF-BB, thrombin, or thrombin receptor-activating peptide blocked hypertrophy in response to serum deprivation.ConclusionsWe conclude that increased α-SMA expression in mesangial cells reflects cellular hypertrophy rather than hyperplasia

    PRELIMINARY STUDIES ON THE BEHAVIOURAL EFFECTS OF THE METHANOL EXTRACT OF LEONOTIS NEPETIFOLIA LINN STEM IN MICE

    Get PDF
    Background: Leonotis nepetifolia Linn (Lamiaceae) is used in traditional medicine for its calming (tranquilizing) effects. The aim of this study was to determine whether there is any scientific justification for this use. To achieve this purpose, we investigated the behavioural effects of the methanol extract of Leonotis nepetifolia stem (37.5, 75 and 150 mg/kg) in mice. Methods: Acute toxicity studies were carried out on the methanol stem extract of Leonotis nepetifolia to determine the LD50. The behavioural tests employed were diazepam-induced sleep onset and duration, hole board assay for exploratory activity, mouse beam walk assay for motor coordination, and the staircase test for the detection of anxiolytic compounds. Preliminary phytochemical screening was also carried out on the extract. Results: The intraperitoneal LD50 value was found to be 3.8 g/kg. The results showed that the extract significantly prolonged the duration of diazepam-induced sleep at the highest dose (150 mg/kg). There was no observable effect on exploratory activity and motor coordination at the doses tested (37.5, 75 and 150 mg/kg). The extract, however, at 150 mg/kg elicited a significant decrease in the number of rearings in the staircase test, an effect also observed in the group of mice injected with an anxiolytic dose of diazepam. The preliminary phytochemical screening revealed the presence of alkaloids, saponins, glycosides and triterpenoids. Conclusion: The results obtained suggest that the crude methanol extract of Leonotis nepetifolia stem possesses some biologically active constituents with potential anxiolytic activity and thus may justify its traditional use as a tranquilize

    Protein Kinase C-α Mediated Downregulation of Low-Density Lipoprotein Receptor-related Protein Expression Increases Urokinase Secretion and Astrocytoma Invasion

    No full text
    Glioblastoma multiforme (GBM) is the most malignant astrocytoma; is characterized by uncontrolled, aggressive cell proliferation and infiltrative growth within the brain; and is resistant to conventional therapy. The molecular and cellular mechanisms governing astrocytic tumor invasion remains poorly understood. We determined the role of low-density lipoprotein receptor-related protein (LRP) in glioblastoma invasive growth. In this study, we demonstrated that activation of protein kinase C (PKC) in astrocytic cell cultures downregulated the expression of LRP and increased the secretion of urokinase (uPA) into conditioned medium. Pretreatment of cell cultures with PKC inhibitors (BIM, Gö 6976) and PI3-kinase inhibitor (LY 294002) and gene silencing with PKCa siRNA abrogated PMAinduced downregulation of LRP, decreased the level of expression of uPA, and inhibited astrocytic tumor cell invasion. Confocal microscopy studies revealed the co-localization of PKC-a and LRP in glioblastoma cell lines. In a Boyden Chamber invasion assay, LRP-deficient glioblastoma cells were more invasive (40%) than LRP-expressing cells, whereas uPA-deficient GBM cells had decreased invasive capacity. Our data show that LRP expression inversely correlates with uPA secretion and GBM invasion. Taken together, our data strongly suggest the involvement of PKCa/PI3 kinase signaling pathways in the regulation of LRP-mediated astrocytoma invasion

    The Protein Kinase C-η Isoform Induces Proliferation in Glioblastoma Cell Lines Through an ERK/Elk-1 Pathway

    No full text
    Glioblastoma multiforme (GBM) is the highest grade of astrocytoma. GBM pathogenesis has been linked to receptor tyrosine kinases and kinases further down signal-transduction pathways – in particular, members of the protein kinase C (PKC) family. The expression and activity of various PKC isoforms are increased in malignant astrocytomas, but not in non-neoplastic astrocytes. This suggests that PKC activity contributes to tumor progression. The level of PKC-η expressed correlates with the degree of phorbol-12-myristate-13-acetate (PMA)-induced proliferation of two glioblastoma cell lines, U-1242 MG and U-251 MG. Normally, U-1242 cells do not express PKC-η, and PMA inhibits their proliferation. Conversely, PMA increases proliferation of U-1242 cells that are stably transfected with PKC-η (U-1242-PKC-η). PMA treatment also stimulates proliferation of U-251 cells, which express PKC-η. Here, we determined that extracellular signal-regulated kinase (ERK) and Elk-1 are downstream targets of PKC-η. Elk-1-mediated transcriptional activity correlates with the PKC-η-mediated mitogenic response. Pretreatment of U-1242-PKC-η cells with inhibitors of PKC or MAPK/ERK kinase (MEK) (bisindolyl maleimide (BIM) or U0126, respectively) blocked both PMA-induced Elk-1 transcriptional activity and PMA-stimulated proliferation. An overexpressed dominant-negative PKC-η reduced the mitogenic response in U-251 cells, as did reduction of Elk-1 by small interfering RNA. Taken together, these results strongly suggest that PKC-η-mediated glioblastoma proliferation involves MEK/mitogen-activated protein (MAP) kinase phosphorylation, activation of ERK and subsequently of Elk-1. Elk-1 target genes involved in GBM proliferative responses have yet to be identified

    Sorafenib tosylate as a radiosensitizer in malignant astrocytoma

    No full text
    Progress in research on the molecular aspects of glioblastoma has yet to provide a medical therapy that significantly improves prognosis. Glioblastoma invariably progress through current treatment regimens with radiotherapy as a key component. Activation of several signaling pathways is thought to be associated with this resistance to radiotherapy. Ras activity is exceptionally high in glioblastoma and may regulate sensitivity to radiotherapy. Raf-1, a downstream effector of Ras, demonstrates a high amount of activity inglioblastoma. Therefore, Raf-1 inhibition should be considered as a mechanism to increase the effectiveness of radiotherapy in treatment regimen. In vitro analysis was performed with a novel Raf-1kinase inhibitor (BAY 54-9085) in culture with the glioblastoma cell line U1242. The cell line was treated in serum-containing media and analyzed for the effect of the BAY 54-9085 alone and BAY 54-9085 combined with radiation on cell death. BAY 54-9085 displayed a cytocidal effect on glioblastoma cells following a 3 day incubation with the drug in serum-containing media. A dose of 2.5 μM displayed moderate cell death which significantly increased with a dose of 5.0 μM. In addition, glioblastoma cells treated with both the BAY 54-9085 and gamma radiation displayed a significant increase in cell death (85.5%) as compared to either BAY 54-9085 (73.1%) or radiation (34.4%) alone. Radiation therapy is a key component of treatment for glioblastoma. A novel Raf-1 inhibitor displayed in vitro evidence of synergistically increasing cell death of glioblastoma cells in combination with radiation

    Activities of Some Medicinal Plants on the Proliferation and Invasion of Brain Tumor Cell Lines

    No full text
    Cancer is a debilitating disease that is on the increase in both developed and developing countries. Anticancer drugs are often expensive, have narrow spectrum of activities, and are associated with toxicities and side effects such as myelosuppression, immunosuppression, gastrointestinal disturbance, alopecia, skin toxicity, and hepatotoxicity. Plants have been the major source of anticancer drugs both in orthodox and traditional medicine. Many of the plants claimed by the traditional medicine practitioners (TMPs) to be effective in the treatment of cancer are yet to be evaluated scientifically. In this work, five medicinal plants used by TMPs in Borno State, Nigeria, were tested against two brain tumor cell lines. Ethanol extracts of Securidaca longepedunculata, Andira inermis subsp. rooseveltii, Annona senegalensis, Carissa edulis, and Parinari polyandra were used. U87 and U231 brain tumor cell lines were used for proliferation assay, U251 cell line was used for the invasion assay in collagen V coated inserts, and U87 cell line was used for the western blot detection of cleaved Poly-ADP-Ribose-Polymerase (PARP). The result revealed that all tested extracts significantly (p<0.05) inhibited the proliferation of U87 and U231 cell lines with the respective IC50 values ranging between 8 and 20 μg/ml for S. longepedunculata and 100 and 90 μg/ml for P. polyandra. The five extracts significantly (p<0.05) inhibited the invasion of U251 cell line at the concentration of 10 μg/ml (S. longepedunculata), 20 μg/ml (A. inermis), 50 μg/ml (A. senegalensis), 50 μg/ml (C. edulis), and 50 μg/ml (P. polyandra). Securidaca longepedunculata extract induced the cleavage of PARP. It was concluded that these medicinal plants have antiproliferative and anti-invasive activities and possess good prospects as source of anticancer agents especially S. longepedunculata which induced apoptosis in U87 cell line

    Phorbol 12-Myristate 13-Acetate Induces Epidermal Growth Factor Receptor Transactivation via Protein Kinase Cdelta/c-Src Pathways in Glioblastoma Cells

    No full text
    Both the epidermal growth factor receptor (EGFR) and protein kinase C (PKC) play important roles in glioblastoma invasive growth; however, the interaction between the EGFR and PKC is not well characterized in glioblastomas. Treatment with EGF stimulated global phosphorylation of the EGFR at Tyr845, Tyr992, Tyr1068, and Tyr1045 in glioblastoma cell lines (U-1242 MG and U-87 MG). Interestingly, phorbol 12-myristate 13-acetate (PMA) stimulated phosphorylation of the EGFR only at Tyr1068 in the two glioblastoma cell lines. Phosphorylation of the EGFR at Tyr1068 was not detected in normal human astrocytes treated with the phorbol ester. PMA-induced phosphorylation of the EGFR at Tyr1068 was blocked by bisindolylmaleimide (BIM), a PKC inhibitor, and rottlerin, a PKCδ-specific inhibitor. In contrast, Gö 6976, an inhibitor of classical PKC isozymes, had no effect on PMA-induced EGFR phosphorylation. Furthermore, gene silencing with PKCδ small interfering RNA (siRNA), siRNA against c-Src, and mutant c-Src(S12C/S48A) and treatment with a c-Src inhibitor (4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d]pyrimidine) abrogated PMA-induced EGFR phosphorylation at Tyr1068. PMA induced serine/threonine phosphorylation of Src, which was blocked by both BIM and rottlerin. Inhibition of the EGFR with AG 1478 did not significantly alter PMA-induced EGFR Tyr1068 phosphorylation, but completely blocked EGF-induced phosphorylation of the EGFR. The effects of PMA on MAPK phosphorylation and glioblastoma cell proliferation were reduced by BIM, rottlerin, the MEK inhibitor U0126, and PKCδ and c-Src siRNAs. Taken together, our data demonstrate that PMA transactivates the EGFR and increases cell proliferation by activating the PKCδ/c-Src pathway in glioblastomas

    Epidermal Growth Factor Receptor-Mediated Regulation of Urokinase Plasminogen Activator Expression and Glioblastoma Invasion via C-SRC/MAPK/AP-1 Signaling Pathways

    No full text
    One of the major pathophysiological features of malignant astrocytomas is their ability to infiltrate surrounding brain tissue. The epidermal growth factor receptor (EGFR) and proteases are known to be overexpressed in glioblastomas (GBMs), but the interaction between the activation of the EGFR and urokinase plasminogen activator (uPA) in promoting astrocytic tumor invasion has not been fully elucidated. Here, we characterized the signal transduction pathway(s) by which EGF regulates uPA expression and promotes astrocytoma invasion. We show that EGFR activation and constitutively active EGFR vIII in GBM cell lines upregulate uPA expression. Small-molecule inhibitors of mitogen-activated protein kinase, tyrosine kinase, and small interfering RNA targeting c-Src blocked uPA upregulation. Similarly, mutations in the activator protein 1 binding site of the uPA promoter reduced EGF-induced increases in uPA promoter activity. Treatment of GBM cells with EGF increased in vitro cell invasion, and the invasive phenotype was attenuated by gene silencing of uPA using small interfering RNA and short hairpin RNA. In addition, uPA knockdown clones formed smaller well-circumscribed tumors than nontarget U1242 control cells in a xenograft GBM mouse model in vivo. In summary, these results suggest that c-Src, mitogen-activated protein kinase, and a composite activator protein 1 on the uPA promoter are responsible for EGF-induced uPA expression and GBM invasion
    corecore