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










Publication year range
1.
Cancer Rep (Hoboken) ; 7(5): e2051, 2024 May.
Article in English | MEDLINE | ID: mdl-38702989

ABSTRACT

BACKGROUND: Glioblastomas are characterized by aggressive behavior. Surgery, radiotherapy, and alkylating agents, including temozolomide are the most common treatment options for glioblastoma. Often, conventional therapies fail to treat these tumors since they develop drug resistance. There is a need for newer agents to combat this deadly tumor. Natural products such as gedunin have shown efficacy in several human diseases. A comprehensive study of gedunin, an heat shock protein (HSP)90 inhibitor, has not been thoroughly investigated in glioblastoma cell lines with different genetic modifications. AIMS: A key objective of this study was to determine how gedunin affects the biological and signaling mechanisms in glioblastoma cells, and to determine how those mechanisms affect the proliferation and apoptosis of glioblastoma cells. METHODS: The viability potentials of gedunin were tested using MTT, cell counts, and wound healing assays. Gedunin's effects on glioma cells were further validated using LDH and colony formation assays. In addition, we investigated the survival and apoptotic molecular signaling targets perturbed by gedunin using Western blot analysis and flow cytometry. RESULTS: Our results show that there was a reduction in cell viability and inhibition of wound healing in the cells tested. Western blot analysis of the gene expression data revealed genes such as EGFR and mTOR/Akt/NF kappa B to be associated with gedunin sensitivity. Gedunin treatment induced apoptosis by cleaving poly ADP-ribose polymerase, activating caspases, and downregulating BCL-xL. Based on these results, gedunin suppressed cell growth and HSP client proteins, resulting in apoptosis in glioblastoma cell lines. CONCLUSION: Our data provide in vitro support for the anticancer activity of gedunin in glioma cells by downregulating cancer survival proteins.


Subject(s)
Apoptosis , Cell Proliferation , Glioblastoma , Limonins , Humans , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Signal Transduction/drug effects , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Survival/drug effects , Antineoplastic Agents/pharmacology
2.
Front Pharmacol ; 13: 952169, 2022.
Article in English | MEDLINE | ID: mdl-36199696

ABSTRACT

The epidermal growth factor (EGFR) receptor is frequently overexpressed in glioblastoma multiforme IV (GBM). Increased expression of EGFR leads to increased proliferation, decreased apoptosis, and increased resistance to chemotherapeutic agents. A small molecule called erlotinib inhibits EGFR receptors by binding to their adenosine triphosphate (ATP) binding sites. It is FDA approved to treat a variety of EGFR-mediated cancers. Several clinical trials have explored a combination of erlotinib with other agents to treat glioblastoma since it is believed that erlotinib would benefit patients with GBM with EGFR mutations or expression. Luteolin, a natural flavonoid, inhibits cell growth and induces apoptosis in cancer cells. We investigated the combined effects of erlotinib and luteolin on proliferation and apoptosis on glioblastoma cell lines overexpressing EGFR or glioma cells expressing truncated EGFR (ΔEGFR). In a concentration-dependent fashion, the combination of luteolin and erlotinib reduced cell proliferation (p < 0.05) and induced apoptosis by cleaving PARP and increasing caspase expression. In addition, the combination of luteolin and erlotinib reduced the phosphorylation of downstream EGFR cell signaling molecules such as Akt, NF kappa B, and STAT3 in a concentration-dependent manner. These findings suggest that combining luteolin with erlotinib offers a potential treatment strategy for glioblastoma multiforme IV.

3.
Cancer Rep (Hoboken) ; 5(11): e1720, 2022 11.
Article in English | MEDLINE | ID: mdl-36195576

ABSTRACT

Biologicals have become an integral part of cancer treatment both as therapeutic agents and as supportive care agents. It is important to know that biologics are large, complex molecular entities requiring extensive immunogenicity testing and pharmacovigilance strategies to ensure no immune response is evoked in the body. Oncology's pharmacological market is dominated by biologics; however, their high development and manufacturing costs are burdensome to health care systems. Biologics being the most expensive prescription drugs on the market limit the accessibility for necessary treatment in the case of many patients. As biologics patents expire, the development of biosimilars is underway in an effort to lower costs and enable patients to access new cancer therapies. Regulatory guidelines for biosimilars have now been established and are constantly being revised to address any issues, facilitating their robust development. Moreover, many scientific societies offer guidance to help stakeholders better understand current regulations and biosimilar's safety. Despite the potential cost benefits, lack of knowledge about biosimilars, and the possibility of immunogenicity have created an uncertain environment for healthcare professionals and patients. In this review, we provide an overview of relevant legislation and regulations, pharmacoeconomics, and stakeholder perceptions regarding biosimilars. The article also describes biosimilars in development, as well as the ones currently available on the market.


Subject(s)
Antineoplastic Agents , Biosimilar Pharmaceuticals , Humans , Biosimilar Pharmaceuticals/therapeutic use , Antineoplastic Agents/adverse effects , Medical Oncology
4.
Anticancer Agents Med Chem ; 21(18): 2512-2519, 2021.
Article in English | MEDLINE | ID: mdl-33622229

ABSTRACT

BACKGROUND: Medicinal plants serve as sources of compounds used to treat other types of cancers. The root of the plant Lophira alata (Ochnaceae) has been used as a component of traditional herbal decoctions administered to cancer patients in southwestern Nigeria. However, the mechanism of the cytotoxic effects of Lophira alata alone or in the presence of phorbol ester has not been investigated in brain tumor cells. OBJECTIVE: This study aimed to examine the cytotoxic potential of the methanolic fraction of Lophira alata root on malignant glioma invasive cellular growth and survival. METHODS: The methanolic fraction of Lophira alata (LAM) was subjected to high-performance liquid chromatography to determine the fingerprints of the active molecules. The antiproliferative effects of Lophira alata were assessed using the MTT and LDH assays. Protein immunoblots were carried out to test the effects of Lophira alata, alone or in the presence of phorbol ester, on survival signaling pathways, such as Akt, mTOR, and apoptotic markers such as PARP and caspases. RESULTS: The methanolic fraction of Lophira alata (LAM) induced a concentration-dependent and time-dependent decrease in glioma cell proliferation. In addition, LAM attenuated phorbol ester-mediated signaling of downstream targets such as Akt/mTOR. Gene silencing using siRNA targeting PKC-alpha attenuated LAM-mediated downregulation of Akt. In addition, LAM induced both PARP and caspase cleavages. The HPLC fingerprint of the fraction indicates the presence of flavonoids. CONCLUSION: LAM decreases cell proliferation and induces apoptosis in glioma cell lines and thus could serve as a therapeutic molecule in the management of gliomas.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Glioblastoma/drug therapy , Ochnaceae/chemistry , Plant Extracts/pharmacology , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Phorbol Esters/pharmacology , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured
5.
Basic Clin Pharmacol Toxicol ; 123(6): 678-686, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29935053

ABSTRACT

Glioblastomas are a subtype of gliomas, which are the most aggressive and deadly form of brain tumours. The epidermal growth factor receptor (EGFR) is over-expressed and amplified in glioblastomas. Luteolin is a common bioflavonoid found in a variety of fruits and vegetables. The aim of this study was to explore the molecular and biological effects of luteolin on EGF-induced cell proliferation and the potential of luteolin to induce apoptosis in glioblastoma cells. In vitro cell viability assays demonstrated that luteolin decreased cell proliferation in the presence or absence of EGF. Immunoblots revealed that luteolin decreased the protein expression levels of phosphorylated Akt, mTOR, p70S6K and MAPK in the presence of EGF. Furthermore, our results revealed the ability of luteolin to induce caspase and PARP cleavages in glioblastoma cells in addition to promoting cell cycle arrest. Our results demonstrated that luteolin has an inhibitory effect on downstream signalling molecules activated by EGFR, particularly the Akt and MAPK signalling pathways, and provided a rationale for further clinical investigation into the use of luteolin as a therapeutic molecule in the management of glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Glioblastoma/drug therapy , Luteolin/pharmacology , Blotting, Western , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Flow Cytometry , Humans
6.
J Pharm Pharmacol ; 69(7): 907-916, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28349530

ABSTRACT

OBJECTIVES: Glioblastoma (GBM) is highly proliferative, infiltrative, malignant and the most deadly form of brain tumour. The epidermal growth factor receptor (EGFR) is overexpressed, amplified and mutated in GBM and has been shown to play key and important roles in the proliferation, growth and survival of this tumour. The goal of our study was to investigate the antiproliferative, apoptotic and molecular effects of apigenin in GBM. METHODS: Proliferation and viability tests were carried out using the trypan blue exclusion, MTT and lactate dehydrogenase (LDH) assays. Flow cytometry was used to examine the effects of apigenin on the cell cycle check-points. In addition, we determined the effects of apigenin on EGFR-mediated signalling pathways by Western blot analyses. KEY FINDINGS: Our results showed that apigenin reduced cell viability and proliferation in a dose- and time-dependent manner while increasing cytotoxicity in GBM cells. Treatment with apigenin-induced is poly ADP-ribose polymerase (PARP) cleavage and caused cell cycle arrest at the G2M checkpoint. Furthermore, our data revealed that apigenin inhibited EGFR-mediated phosphorylation of mitogen-activated protein kinase (MAPK), AKT and mammalian target of rapamycin (mTOR) signalling pathways and attenuated the expression of Bcl-xL. CONCLUSION: Our results demonstrated that apigenin has potent inhibitory effects on pathways involved in GBM proliferation and survival and could potentially be used as a therapeutic agent for GBM.


Subject(s)
Apigenin/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , ErbB Receptors/genetics , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects
7.
J Clin Neurosci ; 21(1): 131-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24139873

ABSTRACT

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 in glioblastoma. 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-1 kinase 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.


Subject(s)
Astrocytoma/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Radiation-Sensitizing Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Flow Cytometry , Humans , Niacinamide/pharmacology , Sorafenib
8.
Turk Neurosurg ; 20(3): 277-85, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20669099

ABSTRACT

AIM: Protein kinase-C (PKC) and NF-kappaB are involved in cell survival, proliferation, migration and radioresistance in glioblastoma multiforme (GBM). We sought to determine the interaction between PKC and NF-kappaB pathways. MATERIAL AND METHODS: The activation of NF-kappaB by PKC alpha and PKC delta was assessed by Western blotting after the stimulation with Phorbol 12- Myristate 13-Acetate (PMA). Gene silencing of PKC alpha , PKC delta and NFkappaB/ p65 with siRNA interference was utilized to evaluate their roles in NFkB activation and cell proliferation. RESULTS: PMA induced the phosphorylation of NF-kappaB/p65 by PKC alpha. Gene silencing with siRNA against NF-kappaB/p65 inhibited [3H]-thymidine incorporation in U1242 GBM cells. PKC delta decelerated the nuclear translocation of activated NF-kappaB/p65 up to 4 hours after the stimulation. PMA induced death was not observed in PKC delta silenced cells where activated NF-kappaB/p65 was located immediately in the nucleus. CONCLUSION: NF-kappaB/p65 is pro-survival and proliferative factor in U1242 GBM cells. PKC alpha is needed to phosphorylate NF-kappaB/p65. PKC delta delays the translocation of active NF-kappaB/p65 into the nucleus. PMA-induced cell death occurred if the phospho-NF-kappaB/p65 was prohibited from entering the nucleus in PKC delta positive cells. Translocation of phosphorylated form of NF-kappaB into the nucleus is critical in GBM cell proliferation.


Subject(s)
Glioblastoma/metabolism , NF-kappa B/metabolism , Protein Kinase C-alpha/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Maleimides/pharmacology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tyrosine/metabolism
9.
Neuro Oncol ; 12(11): 1102-12, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20667897

ABSTRACT

Emerging studies have identified microRNAs (miRNAs) as possible therapeutic tools for the treatment of glioma, the most aggressive brain tumor. Their important targets in this tumor are not well understood. We recently found that the Notch pathway is a target of miRNA-326. Ectopic expression of miRNA-326 in glioma and glioma stem cells induced their apoptosis and reduced their metabolic activity. Computational target gene prediction revealed pyruvate kinase type M2 (PKM2) as another target of miRNA-326. PKM2 has recently been shown to play a key role in cancer cell metabolism. To investigate whether it might be a functionally important target of miR-326, we used RNA interference to knockdown PKM2 expression in glioma cells. Transfection of the established glioma and glioma stem cells with PKM2 siRNA reduced their growth, cellular invasion, metabolic activity, ATP and glutathione levels, and activated AMP-activated protein kinase. The cytotoxic effects exhibited by PKM2 knockdown in glioma and glioma stem cells were not observed in transformed human astrocytes. Western blot analysis of human glioblastoma specimens showed high levels of PKM2 protein, but none was observed in normal brain samples. Strikingly, cells with high levels of PKM2 expressed lower levels of miR-326, suggestive of endogenous regulation of PKM2 by miR-326. Our data suggest PKM2 inhibition as a therapy for glioblastoma, with the potential for minimal toxicity to the brain.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , MicroRNAs/genetics , Pyruvate Kinase/genetics , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/genetics , Gene Expression , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/metabolism , Mutagenesis, Site-Directed , Pyruvate Kinase/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
J Neuropathol Exp Neurol ; 69(6): 582-92, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20467333

ABSTRACT

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.


Subject(s)
Cell Movement/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Glioblastoma/metabolism , MAP Kinase Signaling System/drug effects , Transcription Factor AP-1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Northern , Blotting, Western , Cell Line , Cells, Cultured , ErbB Receptors/genetics , Humans , Image Processing, Computer-Assisted , MAP Kinase Signaling System/physiology , Magnetic Resonance Imaging , Mice , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Time Factors , Transcription Factor AP-1/genetics , Transfection , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/genetics
11.
J Neuropathol Exp Neurol ; 68(5): 535-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19404096

ABSTRACT

Beta-Arrestins act as signal terminators for G protein-coupled receptors; they have also been implicated as scaffolding proteins for Src and mitogen-activated protein kinase signaling pathways and transactivators of receptor tyrosine kinases, suggesting their possible role in development and oncogenic signaling. Dephosphorylation of serine 412 is necessary for Src and mitogen-activated protein kinase transactivation. We hypothesized that altered beta-arrestin 1 phosphorylation and activation status could play a role in gliomagenesis. Using monoclonal anti-phospho-(serine 412)- and total beta-arrestin 1 antibodies, we performed immunohistochemistry on 126 human glioma samples and 7 nonneoplastic controls and Western blot analysis on 5 glioblastomas and 5 nonneoplastic controls. We found high constitutive beta-arrestin 1 phosphorylation in nonneoplastic brain tissue, particularly in neurons and neuropil. Most Grade II and III gliomas retained high beta-arrestin 1 phosphorylation. By contrast, most of the glioblastoma samples (58/81) showed nearly complete beta-arrestin 1 dephosphorylation by immunohistochemistry and decreased relative phosphorylation by Western blot. Expression of constitutively activated epidermal growth factor receptor vIII in U251 cells caused decreased beta-arrestin 1 phosphorylation without altering total beta-arrestin 1 levels. These results suggest that beta-arrestin 1 dephosphorylation/inactivation is associated with aspects of the malignant behavior of glioblastomas.


Subject(s)
Arrestins/metabolism , Glioblastoma/metabolism , Cell Line, Tumor , Follow-Up Studies , Glioblastoma/mortality , Glioblastoma/pathology , Green Fluorescent Proteins/genetics , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Neurons/pathology , Phosphorylation , Retrospective Studies , Serine/metabolism , Transfection , beta-Arrestin 1 , beta-Arrestins
12.
Brain Res Bull ; 78(6): 276-82, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19111909

ABSTRACT

Preparations of Ficus platyphylla have been used in Nigerian traditional medicine for the management of epilepsy for many years and their efficacy is widely acclaimed among the Hausa communities of northern Nigeria. The anticonvulsant properties of the saponin rich fraction (SFG) obtained from the methanol extract of F. platyphylla stem bark were studied on pentylenetetrazole-, strychnine- and maximal electroshock seizures in mice. Effects of SFG were also examined in murine models for neurological disease and on relevant in vitro targets for anticonvulsant drugs. SFG protected mice against pentylenetetrazole- and strychnine-induced seizures; and significantly delayed the onset of myoclonic jerks and tonic seizures. SFG failed to protect mice against maximal electroshock seizures at doses tested. SFG neither abolished the spontaneous discharges induced by 4-aminopyridine in a neonatal rat brain slice model of tonic-clonic epilepsy nor could it modulate chloride currents through GABA(A) receptor channel complex in cultured cortical cells. However, it was able to non-selectively suppress excitatory and inhibitory synaptic traffic, blocked sustained repetitive firing (SRF) and spontaneous action potential firing in these cultured cells. Our results provide scientific evidence that F. platyphylla stem bark may contain psychoactive principles with potential anticonvulsant properties. SFG impaired membrane excitability; a property shared by most anticonvulsants particularly the voltage-gated sodium channel (VGSC) blocking drugs, thus supporting the isolation and development of the saponin components of this plant as anticonvulsant agents.


Subject(s)
Anticonvulsants/therapeutic use , Ficus/chemistry , Phytotherapy , Plant Bark/chemistry , Saponins/therapeutic use , Seizures/drug therapy , 4-Aminopyridine/pharmacology , Animals , Brain/drug effects , Cells, Cultured , Electroshock , Female , Male , Membrane Potentials/drug effects , Mice , Neurons/drug effects , Pentylenetetrazole/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Saponins/isolation & purification , Seizures/chemically induced , Strychnine/pharmacology
13.
Glia ; 56(8): 917-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18383343

ABSTRACT

Previous study reported that the activation of Ras pathway cooperated with E6/E7-mediated inactivation of p53/pRb to transform immortalized normal human astrocytes (NHA/hTERT) into intracranial tumors strongly resembling human astrocytomas. The mechanism of how H-Ras contributes to astrocytoma formation is unclear. Using genetically modified NHA cells (E6/E7/hTERT and E6/E7/hTERT/Ras cells) as models, we investigated the mechanism of Ras-induced tumorigenesis. The overexpression of constitutively active H-RasV12 in E6/E7/hTERT cells robustly increased the levels of urokinase plasminogen activator (uPA) mRNA, protein, activity and invasive capacity of the E6/E7/hTERT/Ras cells. However, the expressions of MMP-9 and MMP-2 did not significantly change in the E6/E7/hTERT and E6/E7/hTERT/Ras cells. Furthermore, E6/E7/hTERT/Ras cells also displayed higher level of uPA activity and were more invasive than E6/E7/hTERT cells in 3D culture, and formed an intracranial tumor mass in a NOD-SCID mouse model. uPA specific inhibitor (B428) and uPA neutralizing antibody decreased uPA activity and invasion in E6/E7/hTERT/Ras cells. uPA-deficient U-1242 glioblastoma cells were less invasive in vitro and exhibited reduced tumor growth and infiltration into normal brain in xenograft mouse model. Inhibitors of Ras (FTA), Raf (Bay 54-9085) and MEK (UO126), but not of phosphatidylinositol 3-kinase (PI3K) (LY294002) and of protein kinase C (BIM) pathways, inhibited uPA activity and cell invasion. Our results suggest that H-Ras increased uPA expression and activity via the Ras/Raf/MEK signaling pathway leading to enhanced cell invasion and this may contribute to increased invasive growth properties of astrocytomas.


Subject(s)
Astrocytes/physiology , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/physiology , Urokinase-Type Plasminogen Activator/metabolism , Analysis of Variance , Animals , Brain Neoplasms/pathology , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/physiology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Transfection/methods , ras Proteins/metabolism
14.
Pituitary ; 11(1): 37-48, 2008.
Article in English | MEDLINE | ID: mdl-17768685

ABSTRACT

OBJECT: We analyzed MMP-9 expression using mRNA and protein level determinations and explored the possibility that matrix metalloproteinase-9 (MMP-9) is a potential biological marker of pituitary adenoma invasiveness and whether MMP-9 could be used to discriminate the extent of invasiveness among different hormonal subtypes, tumor sizes, growth characteristics, and primary versus recurrent tumors. MATERIALS AND METHODS: 73 pituitary tumor specimens were snap frozen in liquid nitrogen immediately after surgical resection. RNA and protein were extracted. MMP-9 mRNA transcripts were analyzed by quantitative RT-PCR. MMP-9 protein activity was analyzed by gelatin zymography and validated by western blot analysis. Immunohistochemistry was performed to identify the presence and localization of MMP-9 in pituitary adenomas. Statistical differences between results were determined using Student's t-test or one way ANOVA. RESULTS: Comparing different hormonal subtypes of noninvasive and invasive pituitary tumors, MMP-9 mRNA expression was significantly increased in the majority of invasive adenomas. Considering the protein levels, our data also showed a significant increase in MMP-9 activity in the majority of invasive adenomas and these differences were confirmed by western blot analysis and immunohistochemistry. In addition, consistent differences in MMP-9 expression levels were found according to tumor subtype, tumor size, tumor extension and primary versus redo-surgery. CONCLUSIONS: MMP-9 expression can consistently distinguish invasive pituitary tumors from noninvasive pituitary tumors and would reflect the extent of invasiveness in pituitary tumors according to tumor subtype, size, tumor extension, primary and redo surgery, even at early stages of invasiveness. MMP-9 may be considered a potential biomarker to determine and predict the invasive nature of pituitary tumors.


Subject(s)
Adenoma/enzymology , Biomarkers, Tumor/analysis , Matrix Metalloproteinase 9/analysis , Pituitary Neoplasms/enzymology , Adenoma/genetics , Adenoma/pathology , Adenoma/surgery , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Blotting, Western , Child , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 9/genetics , Middle Aged , Neoplasm Invasiveness , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery , Predictive Value of Tests , RNA, Messenger/analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Young Adult
15.
Cancer Res ; 67(21): 10241-51, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17974965

ABSTRACT

Aggressive and infiltrative invasion is one of the hallmarks of glioblastoma. Low-density lipoprotein receptor-related protein (LRP) is expressed by glioblastoma, but the role of this receptor in astrocytic tumor invasion remains poorly understood. We show that activation of protein kinase C-alpha (PKC-alpha) phosphorylated and down-regulated LRP expression. Pretreatment of tumor cells with PKC inhibitors, phosphoinositide 3-kinase (PI3K) inhibitor, PKC-alpha small interfering RNA (siRNA), and short hairpin RNA abrogated phorbol 12-myristate 13-acetate-induced down-regulation of LRP and inhibited astrocytic tumor invasion in vitro. In xenograft glioblastoma mouse model and in vitro transmembrane invasion assay, LRP-deficient cells, which secreted high levels of urokinase-type plasminogen activator (uPA), invaded extensively the surrounding normal brain tissue, whereas the LRP-overexpressing and uPA-deficient cells did not invade into the surrounding normal brain. siRNA, targeted against uPA in LRP-deficient clones, attenuated their invasive potential. Taken together, our results strongly suggest the involvement of PKC-alpha/PI3K signaling pathways in the regulation of LRP-mediated astrocytoma invasion. Thus, a strategy of combining small molecule inhibitors of PKC-alpha and PI3K could provide a new treatment paradigm for glioblastomas.


Subject(s)
Astrocytoma/pathology , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Protein Kinase C-alpha/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Astrocytoma/therapy , Cell Line, Tumor , Cell Movement , Humans , Immunoprecipitation , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C-alpha/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
16.
Am J Pathol ; 170(1): 356-65, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200207

ABSTRACT

The complete resection of pituitary adenomas (PAs) is unlikely when there is an extensive local dural invasion and given that the molecular mechanisms remain primarily unknown. DNA microarray analysis was performed to identify differentially expressed genes between nonfunctioning invasive and noninvasive PAs. Gene clustering revealed a robust eightfold increase in matrix metalloproteinase (MMP)-9 expression in surgically resected human invasive PAs and in the (nonfunctioning) HP75 human pituitary tumor-derived cell line treated with phorbol-12-myristate-13-acetate; these results were confirmed by real-time polymerase chain reaction, gelatin zymography, reverse transcriptase-polymerase chain reaction, Western blot, immunohistochemistry, and Northern blot analyses. The activation of protein kinase C (PKC) increased both MMP-9 activity and expression, which were blocked by some PKC inhibitors (Gö6976, bisindolylmaleimide, and Rottlerin), PKC-alpha, and PKC-delta small interfering (si)RNAs but not by hispidin (PKC-beta inhibitor). In a transmembrane invasion assay, phorbol-12-myristate-13-acetate (100 nmol/L) increased the number of invaded HP75 cells, a process that was attenuated by PKC inhibitors, MMP-9 antibody, PKC-alpha siRNA, or PKC-delta siRNA. These results demonstrate that MMP-9 and PKC-alpha or PKC-delta may provide putative therapeutic targets for the control of PA dural invasion.


Subject(s)
Adenoma , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness/genetics , Pituitary Neoplasms , Adenoma/enzymology , Adenoma/pathology , Cell Line, Tumor , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinase 9/biosynthesis , Pituitary Neoplasms/enzymology , Pituitary Neoplasms/pathology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyrones/pharmacology , RNA, Small Interfering , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology
17.
J Biol Chem ; 280(9): 7729-38, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15618223

ABSTRACT

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 Tyr(845), Tyr(992), Tyr(1068), and Tyr(1045) 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 Tyr(1068) in the two glioblastoma cell lines. Phosphorylation of the EGFR at Tyr(1068) was not detected in normal human astrocytes treated with the phorbol ester. PMA-induced phosphorylation of the EGFR at Tyr(1068) was blocked by bisindolylmaleimide (BIM), a PKC inhibitor, and rottlerin, a PKCdelta-specific inhibitor. In contrast, Go 6976, an inhibitor of classical PKC isozymes, had no effect on PMA-induced EGFR phosphorylation. Furthermore, gene silencing with PKCdelta 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 Tyr(1068). 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 Tyr(1068) 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 PKCdelta and c-Src siRNAs. Taken together, our data demonstrate that PMA transactivates the EGFR and increases cell proliferation by activating the PKCdelta/c-Src pathway in glioblastomas.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/metabolism , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation , src-Family Kinases/metabolism , Acetophenones/pharmacology , Astrocytes/metabolism , Benzopyrans/pharmacology , Blotting, Western , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Models, Biological , Nitriles/pharmacology , Phosphorylation , Protein Isoforms , Protein Kinase C-delta , RNA, Small Interfering/metabolism , Thymidine/chemistry , Time Factors , Tyrosine/chemistry
18.
Pharmacol Biochem Behav ; 77(4): 751-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15099920

ABSTRACT

The effects of the ethanol extract of Pavetta crassipes on the central nervous system (CNS) and on actions of some selected centrally acting drugs were studied in mice and rats. These studies were carried out using the spontaneous motor activity (SMA), amphetamine-induced hyperactivity and stereotyped behaviour, pentobarbital-induced hypnosis and exploratory activity, apomorphine-induced climbing and haloperidol-induced catalepsy in rats. The results demonstrated that the extract of P. crassipes dose-dependently decreased SMA in mice and attenuated amphetamine-induced hyperactivity and the different episodes of stereotypic behavioural patterns induced by amphetamine. In addition, the extract decreased the number of head dips in the exploratory activity test and potentiated pentobarbital-induced sleeping time in rats. Furthermore, the extract inhibited apomorphine-induced climbing in mice and potentiated haloperidol-induced catalepsy in rats. Our results suggest that the extract of P. crassipes contains biologically active substance(s) that might be acting centrally through the inhibition of dopaminergic pathway or a system linked to this pathway to mediate the observed pharmacological effects.


Subject(s)
Motor Activity/drug effects , Rubiaceae , Stereotyped Behavior/drug effects , Animals , Apomorphine/pharmacology , Dose-Response Relationship, Drug , Female , Male , Mice , Motor Activity/physiology , Plant Extracts/pharmacology , Plant Leaves , Rats , Rats, Wistar , Stereotyped Behavior/physiology
19.
Biol Pharm Bull ; 26(12): 1674-80, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14646169

ABSTRACT

Pavetta crassipes leaf is routinely used locally in Nigeria for the management of respiratory disorders and hypertension. The hypotensive and other cardiovascular effects of Pavetta crassipes were investigated in cats and rats. The effects of the extract on rat and cat blood pressures, isolated rat atria, rat portal vein, isolated rat aorta and rat vas deferens were studied. Specific receptor antagonists (atropine, mepyramine, phentolamine, propranolol) were used to elucidate the underlying mechanism(s) involved in the cardiovascular changes induced by P. crassipes. The results revealed that the ethanolic extract of Pavetta crassipes lowered the blood pressures of cats and rats in a dose dependent manner. The extract also caused a concentration-dependent decrease in the force of contraction of the isolated rat atria and rat portal vein. The decreases in blood pressure values were attenuated in the presence of a beta-adrenoceptor antagonist, propranolol. The extract also attenuated isoprenaline-induced contraction of the rat atria. However, the extract did not affect contractions evoked by KCl, norepinephrine and 5-HT on the rat aorta. Pavetta crassipes contains biologically active substances that may be useful in the management of hypertension.


Subject(s)
Hypertension/drug therapy , Hypotension/chemically induced , Phytotherapy , Plant Extracts/pharmacology , Plant Leaves/chemistry , Rubiaceae/chemistry , Animals , Aorta/drug effects , Atrial Appendage/drug effects , Atrial Appendage/physiology , Blood Pressure/drug effects , Cats , Dose-Response Relationship, Drug , Ethanol , Female , Injections, Intravenous , Male , Medicine, African Traditional , Mice , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Portal Vein/drug effects , Rats , Vas Deferens/drug effects
20.
Pharmacol Toxicol ; 91(2): 71-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12420795

ABSTRACT

Behavioral effects of methyl angolensate were investigated in mice and rats. Spontaneous motor activity, pentobarbital sleeping time, amphetamine-stereotyped behaviour, exploratory activity and apomorphine-induced climbing studies in mice were evaluated. The results revealed that methyl angolensate reduced spontaneous motor activity in mice, prolonged the duration of pentobarbital sleeping time in rats and attenuated amphetamine-induced stereotype behaviour in rats. Methyl angolensate also decreased exploratory activity in mice and reduced the rate of apomorphine-induced climbing in mice at the doses tested. It is suggested that methyl angolensate possesses some biologically active principles that are sedative in nature.


Subject(s)
Behavior, Animal/drug effects , Motor Activity/drug effects , Plant Extracts/toxicity , Sleep/drug effects , Triterpenes/toxicity , Analysis of Variance , Animals , Female , Lethal Dose 50 , Male , Mice , Pentobarbital/pharmacology , Rats , Rats, Wistar , Triterpenes/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL