Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Biochem Cell Biol ; 45(7): 1377-88, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23603149

ABSTRACT

The neuroinflammatory process is thought to contribute to the progression of neurological disorders and brain pathologies. The release of pro-inflammatory cytokines and chemokines by activated glial cells, astrocytes and microglia plays an important role in this process. However, the role of hypoxia-inducible factor-1α (HIF-1α), the key transcription factor regulating the expression of hypoxia-inducible genes, during glial activation is less known. Thus, we examined the significance of HIF-1α in three experimental models: first in an acute model of inflammation induced by pro-inflammatory cytokines TNF-α, IL-1ß and IFN-γ; secondly, in a chronic model of inflammation using an APPswe/PS1dE9 (APP/PS1) transgenic mouse model of Alzheimer's disease and thirdly via the inhibition of the PI3K/AKT pathway in a model of neuronal apoptosis. During acute glial inflammation induced by in vitro administration of TNF-α, IL-1ß and IFN-γ, mRNA expression levels of HIF-1α were significantly upregulated; however, this effect was blocked by SP600126, a pharmacological inhibitor of mitogen-activated protein kinases (MAPKs). These data suggest that MAPKs could be involved in HIF-1α regulation. In addition, we observed that HIF-1α is not involved in the neuronal apoptotic process mediated by PI3-kinase inhibition, which is regulated by c-Jun. Finally, we did not detect significant differences in the expression of HIF-1α mRNA in APP/PS1 mice during the course of the study (3-12 months of age). Thus, we demonstrated that HIF-1α has a prominent role in acute but not in chronic inflammatory processes, such as the one which occurs in the APP/PS1 experimental model of AD. Moreover, HIF-1α is not involved in neuronal apoptosis after PI3K/AKT inhibition.


Subject(s)
Alzheimer Disease/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuroglia , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
2.
Pharmacol Res ; 65(1): 66-73, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21875668

ABSTRACT

In the present study, we evaluated the effects of roscovitine (Rosco) and flavopiridol (Flavo), both of which are classified as cyclin-dependent kinase (CDK) inhibitors, on apoptosis induced by the inhibition of PI3K/AKT pathway in cerebellar granule neurons (CGNs). Our results demonstrate that both CDK inhibitors prevented apoptosis induced by LY294002 (LY), as also occurs with SB415286 (SB4), a selective GSK3ß inhibitor. Our findings also indicate that these CDK inhibitors inhibit GSK3ß, representing a potential pharmacological mechanism involved in their neuroprotective properties. Thus, the increased activity of GSK3ß induced by LY294002 and detected by dephosphorylation at Ser9 was prevented by both compounds. Likewise, GSK3ß activity was measured by a radioactivity assay, revealing that CDK inhibitors and SB415286 prevented the increase in GSK3ß activity induced by PI3K inhibition. In addition, we analysed c-Jun, which is also a mediator of PI3K inhibition-induced apoptosis. However, neither of the CDK inhibitors nor SB415286 prevented the increase in c-Jun phosphorylation induced by PI3K inhibition. Therefore, our data identify GSK3ß as a crucial mediator of CGN apoptosis induced by PI3K inhibition and indicate that the antiapoptotic effects of CDKs are mediated by the inhibition of this pharmacological target.


Subject(s)
Cerebellum/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebellum/enzymology , Cerebellum/pathology , Cyclin-Dependent Kinases/metabolism , Cytoprotection , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Neurons/enzymology , Neurons/pathology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Roscovitine , Signal Transduction/drug effects
3.
Neurochem Int ; 57(7): 774-82, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20817061

ABSTRACT

In the present study the role of JAK/STAT and Akt in apoptosis was evaluated in cerebellar granule cells after treatment with the mitochondrial toxin MPP(+). Firstly, we evaluated the role of the prosurvival Akt pathway in MPP(+)-induced apoptosis and found that MPP(+) rapidly reduced the phosphorylation of Akt at Ser473. Since PTEN is an upstream regulator of Akt, its inhibition with bpV(pic) (1-30 µM) should activate Akt, however, it did not attenuate CGC cell death mediated by MPP(+) but protected CGC from apoptosis mediated by S/K deprivation. We also demonstrated that after the treatment with the complex I inhibitor, the expression levels of STAT1 increased and the levels of STAT3 decreased at the time points tested (0.5-8h). Meanwhile, pharmacological inhibition of the JAK/STAT pathway with AG490 (10-40 µM) was neuroprotective, probably due to its antioxidant properties, the Jak2-inhibitor-II potentiated MPP(+) neurotoxicity. Collectively, our data indicate that the treatment of CGC with the neurotoxin MPP(+) decreased two prosurvival pathways: STAT3 and Akt. Meanwhile Akt activation, using a PTEN inhibitor, did not play a prominent role in neuroprotection; loss of STAT3 could be a signal pathway involved in neuroprotection against the Parkinsonian neurotoxin MPP(+).


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Apoptosis/physiology , Janus Kinase 2/physiology , Neurons/enzymology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebellum/drug effects , Cerebellum/enzymology , Janus Kinase 2/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/physiology , Neurons/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects
4.
Cell Mol Life Sci ; 67(22): 3865-82, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20502937

ABSTRACT

In the present study we demonstrated that neurotoxin MPP(+)-induced DNA damage is followed by ataxia telangiectasia muted (ATM) activation either in cerebellar granule cells (CGC) or in B65 cell line. In CGC, the selective ATM inhibitor KU-55933 showed neuroprotective effects against MPP(+)-induced neuronal cell loss and apoptosis, lending support to the key role of ATM in experimental models of Parkinson's disease. Likewise, we showed that knockdown of ATM levels in neuroblastoma B65 cells using an ATM-specific siRNA attenuates the phosphorylation of retinoblastoma protein without affecting other cell-cycle proteins involved in the G(0)/G(1) cell-cycle phase. Moreover, we demonstrated DNA damage, in human brain samples of PD patients. These findings support a model in which MPP(+) leads to ATM activation with a subsequent DNA damage response and activation of pRb. Therefore, this study demonstrates a new link between DNA damage by MPP(+) and cell-cycle re-entry through retinoblastoma protein phosphorylation.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Cell Cycle Proteins/metabolism , Cerebellum/cytology , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Neurotoxins/pharmacology , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Brain/pathology , Cell Cycle , Cell Line , Cells, Cultured , Female , Humans , Male , Middle Aged , Morpholines/pharmacology , Pyrones/pharmacology , Rats , Rats, Sprague-Dawley
5.
J Cell Biochem ; 110(1): 210-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20213763

ABSTRACT

Ataxia telangiectasia mutated protein (ATM) is a member of the phosphatidylinositol-3 kinase (PI3K) family, which has a role in the cellular response to DNA double-strand breaks (DSBs). In the present study, we evaluated the role of ATM in cell-cycle control in dopaminergic rat neuroblastoma B65 cells. For this purpose, ATM activity was either inhibited pharmacologically with the specific inhibitor KU-55933, or the ATM gene was partially silenced by transfection with small interfering RNA (siRNA). Our data indicate that although ATM inhibition did not affect the cell cycle, both treatments specifically decreased the levels of cyclin A and retinoblastoma protein (pRb), phosphorylated at Ser780. Furthermore, ATM inhibition decreased the active form of p53, which is phosphorylated at Ser15, and also decreased Bax and p21 expression. Using H(2)O(2) as a positive control of DSBs, caused a rapid pRb phosphorylation, this was prevented by KU-55933 and siRNA treatment. Collectively, our data demonstrate how a new molecular network on ATM regulates the cell cycle through the control of pRb phosphorylation. These findings support a new target of ATM.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Line, Tumor , Gene Silencing/drug effects , Hydrogen Peroxide/pharmacology , Molecular Sequence Data , Morpholines/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Pyrones/pharmacology , RNA, Small Interfering/metabolism , Rats , Retinoblastoma Protein/chemistry , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...