Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Signal ; 23(11): 1869-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21756999

ABSTRACT

The insulin-like growth factor (IGF-1) induces hypoxia inducible factor (HIF-1α) regulated genes in glioblastoma multiforme (GBM). As HIF-1α links inflammatory and oncogenic pathways in GBM, we investigated whether IGF-1 affects HIF-1α to regulate inflammatory response in glioma cells under normoxia. IGF-1 induced Ras and Calmodulin-dependent kinase II (CaMKII) regulated HIF-1α transcriptional activity in glioma cells. Increase in HIF-1α was concurrent with decreased Toll-like receptor (TLR9) and CXCR4 expression and elevated suppressor of cytokine signaling (SOCS3) levels. Interestingly, while synthetic CpG containing oligodeoxynucleotide TLR9 agonist (CpG DNA) decreased IGF-1 mediated increase in HIF-1α activity, siRNA mediated knockdown of HIF-1α decreased TLR9 levels. This suggested that IGF-1 induced HIF-1α-TLR9 axis is regulated by both positive and negative feedback loops. Importantly, TLR9 agonist reversed the effect of IGF-1 on CXCR4 and SOCS3 expression. While knockdown of HIF-1α abrogated IGF-1 mediated increase in SOCS3 it elevated IGF-1 induced decrease in CXCR4 levels. Thus HIF-1α positively and negatively regulates SOCS3 and CXCR4 expression respectively, in glioma cells. Though TLR9 agonist had no additive effect on IGF-1 mediated increase in pro-inflammatory cytokines IL-1ß, IL-6 and IL-8, treatment with TLR9 agonist alone elevated expression of these pro-inflammatory cytokines. Our studies indicate that a complex HIF-1α-TLR9 cross-talk sustains a self-regulating cycle of inflammatory response through intrinsic negative and positive feedback mechanisms.


Subject(s)
Feedback, Physiological , Glioblastoma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation/metabolism , Insulin-Like Growth Factor I/metabolism , Signal Transduction/genetics , Toll-Like Receptor 9/metabolism , Blotting, Western , Cell Line, Tumor , Cytokines/analysis , Cytokines/biosynthesis , Female , Gene Expression , Gene Silencing/drug effects , Genes, Reporter , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Insulin-Like Growth Factor I/pharmacology , Luciferases/analysis , Oxygen , Protein Array Analysis , RNA, Small Interfering/pharmacology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Toll-Like Receptor 9/genetics , Transfection , ras Proteins/genetics , ras Proteins/metabolism
2.
J Mol Med (Berl) ; 89(2): 123-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20865400

ABSTRACT

We observed elevated levels of pro-inflammatory cytokine IL-1ß in glioblastoma multiforme tumor samples. Since hypoxia-inducible factor-1α (HIF-1α) plays a crucial role in linking inflammatory and oncogenic pathways, we investigated the effect of IL-1ß on HIF-1α expression in glioma cells under normoxia. IL-1ß-mediated elevation of HIF-1α transcriptional activity was dependent on Ras-induced NF-κB activation, as IL-1ß failed to induce NF-κB and HIF-1α activity in cells transfected with dominant negative RasN17. Increased Ras expression was accompanied by increased phosphorylation of Ras effectors AKT, ERK, JNK, and p38MAPK. While inhibition of these effectors individually failed to block the IL-1ß-mediated increase in HIF-1α induction, co-inhibition of both AKT and ERK resulted in a significant decrease in IL-1ß-induced HIF-1α activation. Interestingly, IL-1ß elevated Wnt-1 expression in a Ras-dependent manner, and small interfering RNA (siRNA)-mediated knockdown of Wnt-1 decreased HIF-1α activity. Although Wnt-1-mediated HIF-1α was independent of the canonical Wnt/ß-catenin signaling pathway, it regulated HIF-1α through NF-κB. siRNA-mediated HIF-1α knockdown attenuated elevated IL-1ß mRNA levels induced upon IL-1ß treatment. This was accompanied by increased interaction of HIF-1α with HIF responsive element on the IL-1ß promoter upon IL-1ß treatment, under normoxia. Our studies highlights for first time that (1) Ras is a key mediator of IL-1ß-induced NF-κB and HIF-1α activation, under normoxia; (2) Wnt-1 regulates IL-1ß-mediated HIF-1α induction via NF-κB; (3) Ras and Wnt-1 are intermediaries in the canonical IL-1ß-NF-κB signaling pathway downstream of MyD88; and (4) IL-1ß-induced HIF-1α drives a HIF-1α-IL-1ß autocrine loop to maintain persistently elevated IL-1ß level.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioblastoma/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Autocrine Communication , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , beta Catenin/metabolism
3.
BMC Cancer ; 10: 328, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20576128

ABSTRACT

BACKGROUND: The highly resistant nature of glioblastoma multiforme (GBM) to chemotherapy prompted us to evaluate the efficacy of bicyclic triterpenoid Iripallidal against GBM in vitro. METHODS: The effect of Iripallidal on proliferation and apoptosis in glioma cell lines was evaluated by MTS, colony formation and caspase-3 activity. The effect of iripallidal to regulate (i) Akt/mTOR and STAT3 signaling (ii) molecules associated with cell cycle and DNA damage was evaluated by Western blot analysis. The effect of Iripallidal on telomerase activity was also determined. RESULTS: Iripallidal (i) induced apoptosis, (ii) inhibited Akt/mTOR and STAT3 signaling, (iii) altered molecules associated with cell cycle and DNA damage, (iv) inhibited telomerase activity and colony forming efficiency of glioma cells. In addition, Iripallidal displayed anti-proliferative activity against non-glioma cancer cell lines of diverse origin. CONCLUSION: The ability of Iripallidal to serve as a dual-inhibitor of Akt/mTOR and STAT3 signaling warrants further investigation into its role as a therapeutic strategy against GBM.


Subject(s)
Acrolein/analogs & derivatives , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cyclohexanols/pharmacology , Glioblastoma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Acrolein/pharmacology , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Telomerase/antagonists & inhibitors , Telomerase/metabolism
4.
J Cell Mol Med ; 14(8): 2151-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19583803

ABSTRACT

The present study identified a novel mechanism of induction of apoptosis in glioblastoma cells by scriptaid - a histone deacetylase (HDAC) inhibitor. Scriptaid reduced glioma cell viability by increasing Jun N-terminal kinase (JNK) activation. Although scriptaid induced activation of both p38MAPK and JNK, it was the inhibition of JNK that attenuated scriptaid-induced apoptosis significantly. Scriptaid also increased the expression of (i) p21 and p27 involved in cell-cycle regulation and (ii) γH2AX associated with DNA damage response in a JNK-dependent manner. Treatment with scriptaid increased Ras activity in glioma cells, and transfection of cells with constitutively active RasV12 further sensitized glioma cells to scriptaid-induced apoptosis. Scriptaid also inhibited telomerase activity independent of JNK. Taken together, our findings indicate that scriptaid (i) induces apoptosis and reduces glioma cell proliferation by elevating JNK activation and (ii) also decreases telomerase activity in a JNK-independent manner.


Subject(s)
Apoptosis/drug effects , Hydroxylamines/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Quinolines/pharmacology , Telomerase/metabolism , Acetylation/drug effects , Anthracenes/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Polymerase Chain Reaction/methods , Signal Transduction/drug effects , Telomerase/antagonists & inhibitors , Telomerase/genetics , ras Proteins/metabolism
5.
Free Radic Biol Med ; 47(4): 364-74, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19409983

ABSTRACT

The poor prognosis of glioblastoma multiforme and lack of effective therapy have necessitated the identification of new treatment strategies. We have previously reported that elevation of oxidative stress induces apoptosis of glioma cells. Because the farnesyltransferase inhibitor manumycin is known to induce reactive oxygen species (ROS) generation, we evaluated the effects of manumycin on glioma cells. Manumycin induced glioma cell apoptosis by elevating ROS generation. Treatment with the ROS inhibitor N-acetylcysteine blocked manumycin-induced apoptosis, caspase-3 activity, and PARP expression, indicating the involvement of increased ROS in the proapoptotic activity of manumycin. This heightened ROS level was accompanied by a concurrent decrease in antioxidants such as superoxide dismutase (SOD-1) and thioredoxin (TRX-1). SOD-1 overexpression protects glioma cells from manumycin-induced apoptosis. In addition, small interfering RNA-mediated knockdown of SOD-1 and TRX-1 expression also increased ROS generation and sensitivity of glioma cells to manumycin-induced cell death. Interestingly, suppressing ROS generation prevented manumycin-induced Ras inhibition. This study reports for the first time that Ras inhibition by manumycin is due to heightened ROS levels. We also report for the first time that manumycin inhibits the phosphorylation of signal transducer and activator of transcription 3 and telomerase activity in a ROS-dependent manner, which plays a crucial role in glioma resistance to apoptosis. In addition manumycin (i) induced the DNA-damage repair response, (ii) affected cell-cycle-regulatory molecules, and (iii) impaired the colony-forming ability of glioma cells in a ROS-dependent manner.


Subject(s)
Glioblastoma/pathology , Glioma/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Telomerase/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Farnesyltranstransferase/antagonists & inhibitors , Gene Expression Regulation/drug effects , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/physiopathology , Glioblastoma/therapy , Glioma/enzymology , Glioma/genetics , Glioma/physiopathology , Glioma/therapy , Humans , Oxidative Stress , Phosphorylation , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polyenes/pharmacology , Polyenes/therapeutic use , Polyunsaturated Alkamides/pharmacology , Polyunsaturated Alkamides/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Thioredoxins/genetics , Thioredoxins/metabolism
6.
Mol Oncol ; 3(1): 77-83, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19383369

ABSTRACT

We investigated the pro-inflammatory response mediated by TNFalpha in glioblastoma and whether treatment with organoselenium Ebselen (2-phenyl-1,2-benzisoselenazol-3[2H]one) can affect TNFalpha induced inflammatory response. Exposure to TNFalpha increased the expression of pro-inflammatory mediator interleukin IL-6, IL-8, monocyte chemoattractant protein-1 (MCP-1) and cyclooxygenase (COX-2). Treatment with Ebselen abrogated TNFalpha induced increase in pro-inflammatory mediators. Ebselen not only abrogated TNFalpha induced enhanced invasiveness of glioma cells by down-regulating matrix metallo proteinase (MMP-9) and urokinase plasminogen (uPa) activity, but also inhibited glioma cell migration. Treatment with Ebselen also down-regulated the enhanced ROS production of TNFalpha treated glioma cells. In addition, Ebselen induced DNA damage repair signaling response in glioma cells both in the presence and absence of TNFalpha. These studies indicate that together with its known ability to sensitize glioma cell to TNFalpha induced apoptosis, Ebselen can overcome TNFalpha induced pro-inflammatory mediators to prevent a build up of a deleterious pro-inflammatory tumor microenvironment.


Subject(s)
Azoles/pharmacology , Glioblastoma/pathology , Inflammation/drug therapy , Organoselenium Compounds/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Azoles/therapeutic use , Cell Movement/drug effects , DNA Repair , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation Mediators/analysis , Isoindoles , Organoselenium Compounds/therapeutic use
7.
J Biosci ; 33(3): 405-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19005240

ABSTRACT

Until recently, the neuroscience community held the belief that glial cells such as astrocytes and oligodendrocytes functioned solely as "support" cells of the brain. In this role, glial cells simply provide physical support and housekeeping functions for the more important cells of the brain, the neurons. However, this view has changed radically in recent years with the discovery of previously unrecognized and surprising functions for this underappreciated cell type. In the past decade or so, emerging evidence has provided new insights into novel glial cell activities such as control of synapse formation and function, communication,cerebrovascular tone regulation, immune regulation and adult neurogenesis. Such advances in knowledge have effectively elevated the role of the astrocyte to one that is more important than previously realized. This review summarizes the past and present knowledge of glial cell functions that has evolved over the years, and has resulted in a new appreciation of astrocytes and their value in studying the neurobiology of human brain cells and their functions. In this review, we highlight recent advances in the role of glial cells in physiology, pathophysiology and, most importantly, in adult neurogenesis and "stemness", with special emphasis on astrocytes.


Subject(s)
Astrocytes/physiology , Neurogenesis , Adult Stem Cells/physiology , Animals , Humans , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/physiology , Receptor Cross-Talk , Synaptic Transmission
8.
J Neurochem ; 107(3): 616-27, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18710416

ABSTRACT

The anti-neoplastic property of alkyl phospholipids has been tested for the treatment of several malignancies. In this study, we evaluated the efficacy of miltefosine (Hexadecylphosphocholine--an alkyl phospholipids analogue) on glioblastoma multiforme. In this study, we demonstrate that miltefosine-induced apoptosis is accompanied by elevated Fas, Fas-associated death domain (FADD) expression, caspase-8 activity and the increased distribution of Fas and FADD towards lipid raft microdomain to form death inducing signaling complex. Treatment with miltefosine resulted in increase in Ras, extracellular signal-regulated kinase (ERK) and p38MAPK activity. Expression of dominant-negative Ras (Ras N17) attenuated miltefosine-mediated apoptosis. Although inhibition of both ERK and p38MAPK decreased the pro-apoptotic effects of miltefosine, it was the inhibition of ERK and not p38MAPK activation that decreased Fas and FADD expression. An ERK-dependent increase in the expression of gammaH2AX-involved in response to DNA double-stranded breaks was also observed. Taken together, our findings suggest the involvement of ERK activation in miltefosine-induced glioma cell apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Glioma/enzymology , Phosphorylcholine/analogs & derivatives , fas Receptor/metabolism , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fas-Associated Death Domain Protein/metabolism , Flow Cytometry , Glioma/pathology , Humans , Immunoprecipitation , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Phosphorylcholine/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism
9.
Mol Med Rep ; 1(4): 511-5, 2008.
Article in English | MEDLINE | ID: mdl-21479441

ABSTRACT

Polyphenol epigallocatechin-3-gallate (EGCG) induced apoptosis in glioma cells by elevating oxidative stress through increased reactive oxygen species (ROS) generation. Signs of apoptosis included altered mitochondrial membrane potential and elevated expression of caspase-3 and cytochrome c. The increase in ROS was concomitant with the decrease in expression of thioredoxin (TRX-1) and ceruloplasmin (CP), mediators associated with protection against oxidative stress. EGCG downregulated the levels of pro-inflammatory cytokine interleukin (IL)-6 and chemokines IL-8, monocyte-chemoattractant protein (MCP)-1 and RANTES. EGCG also decreased the invasive potential of gliomas, possibly by affecting the urokinase plasminogen activator (uPA) and cytoskeletal architecture. Our study indicates that EGCG might serve as an effective therapeutic strategy against glioma as it not only promotes cell death through redox perturbation, but also downregulates the release of pro-inflammatory mediators while concomitantly decreasing the invasive potential of glioma cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...