Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Bioorg Chem ; 146: 107255, 2024 May.
Article in English | MEDLINE | ID: mdl-38457955

ABSTRACT

Monoaminooxidases (MAOs) are important targets for drugs used in the treatment of neurological and psychiatric disorders and particularly on Parkinson's Disease (PD). Compounds containing a trans-stilbenoid skeleton have demonstrated good selective and reversible MAO-B inhibition. Here, twenty-two (Z)-3-benzylidenephthalides (benzalphthalides, BPHs) displaying a trans-stilbenoid skeleton have been synthesised and evaluated as inhibitors of the MAO-A and MAO-B isoforms. Some BPHs have selectively inhibited MAO-B, with IC50 values ranging from sub-nM to µM. The most potent compound with IC50 = 0.6 nM was the 3',4'-dichloro-BPH 16, which showed highly selective and reversible MAO-B inhibitory activity. Furthermore, the most selective BPHs displayed a significant protection against the apoptosis, and mitochondrial toxic effects induced by 6-hydroxydopamine (6OHDA) on SH-SY5Y cells, used as a cellular model of PD. The results of virtual binding studies on the most potent compounds docked in MAO-B and MAO-A were in agreement with the potencies and selectivity indexes found experimentally. Additionally, related to toxicity risks, drug-likeness and ADME properties, the predictions found for the most relevant BPHs in this research were within those ranges established for drug candidates.


Subject(s)
Neuroblastoma , Parkinson Disease , Stilbenes , Humans , Molecular Docking Simulation , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemistry , Parkinson Disease/drug therapy , Phthalic Acids/chemistry , Phthalic Acids/pharmacology , Structure-Activity Relationship , Benzyl Compounds/chemical synthesis , Benzyl Compounds/chemistry , Benzyl Compounds/pharmacology
2.
Int J Mol Sci ; 22(14)2021 Jul 06.
Article in English | MEDLINE | ID: mdl-34298892

ABSTRACT

One of the most important mechanisms of preconditioning-mediated neuroprotection is the attenuation of cell apoptosis, inducing brain tolerance after a subsequent injurious ischemia. In this context, the antiapoptotic PI3K/AKT signaling pathway plays a key role by regulating cell differentiation and survival. Active AKT is known to increase the expression of murine double minute-2 (MDM2), an E3-ubiquitin ligase that destabilizes p53 to promote the survival of cancer cells. In neurons, we recently showed that the MDM2-p53 interaction is potentiated by pharmacological preconditioning, based on subtoxic stimulation of NMDA glutamate receptor, which prevents ischemia-induced neuronal apoptosis. However, whether this mechanism contributes to the neuronal tolerance during ischemic preconditioning (IPC) is unknown. Here, we show that IPC induced PI3K-mediated phosphorylation of AKT at Ser473, which in turn phosphorylated MDM2 at Ser166. This phosphorylation triggered the nuclear stabilization of MDM2, leading to p53 destabilization, thus preventing neuronal apoptosis upon an ischemic insult. Inhibition of the PI3K/AKT pathway with wortmannin or by AKT silencing induced the accumulation of cytosolic MDM2, abrogating IPC-induced neuroprotection. Thus, IPC enhances the activation of PI3K/AKT signaling pathway and promotes neuronal tolerance by controlling the MDM2-p53 interaction. Our findings provide a new mechanistic pathway involved in IPC-induced neuroprotection via modulation of AKT signaling, suggesting that AKT is a potential therapeutic target against ischemic injury.


Subject(s)
Ischemia/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , HEK293 Cells , Humans , Ischemic Preconditioning/methods , Mice , Mice, Inbred C57BL , Neuroprotection/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/physiology , Wortmannin/metabolism
3.
Neurochem Res ; 46(1): 51-63, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32189131

ABSTRACT

A new era for neuroprotective strategies is emerging in ischemia/reperfusion. This has forced to review the studies existing to date based in neuroprotection against oxidative stress, which have undoubtedly contributed to clarify the brain endogenous mechanisms, as well as to identify possible therapeutic targets or biomarkers in stroke and other neurological diseases. The efficacy of exogenous administration of neuroprotective compounds has been shown in different studies so far. However, something must be missing to get these treatments successfully applied in the clinical environment. Here, the mechanisms involved in neuronal protection against physiological level of ROS and the main neuroprotective signaling pathways induced by excitotoxic and ischemic stimuli are reviewed. Also, the endogenous ischemic tolerance in terms of brain self-protection mechanisms against subsequent cerebral ischemia is revisited to highlight how the preconditioning has emerged as a powerful tool to understand these phenomena. A better understanding of endogenous defense against exacerbated ROS and metabolism in nervous cells will therefore aid to design pharmacological antioxidants targeted specifically against oxidative damage induced by ischemic injury, but also might be very valuable for translational medicine.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/therapy , Ischemic Preconditioning , Neuroprotection/physiology , Neuroprotective Agents/therapeutic use , Animals , Astrocytes/metabolism , Gene Expression/physiology , Humans , Neurons/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
4.
Transl Stroke Res ; 10(2): 204-215, 2019 04.
Article in English | MEDLINE | ID: mdl-29687302

ABSTRACT

Cerebral preconditioning (PC) confers endogenous brain protection after stroke. Ischemic stroke patients with a prior transient ischemic attack (TIA) may potentially be in a preconditioned state. Although PC has been associated with the activation of pro-survival signals, the mechanism by which preconditioning confers neuroprotection is not yet fully clarified. Recently, we have described that PC-mediated neuroprotection against ischemic insult is promoted by p53 destabilization, which is mediated by its main regulator MDM2. Moreover, we have previously described that the human Tp53 Arg72Pro single nucleotide polymorphism (SNP) controls susceptibility to ischemia-induced neuronal apoptosis and governs the functional outcome of patients after stroke. Here, we studied the contribution of the human Tp53 Arg72Pro SNP on PC-induced neuroprotection after ischemia. Our results showed that cortical neurons expressing the Pro72-p53 variant exhibited higher PC-mediated neuroprotection as compared with Arg72-p53 neurons. PC prevented ischemia-induced nuclear and cytosolic p53 stabilization in Pro72-p53 neurons. However, PC failed to prevent mitochondrial p53 stabilization, which occurs in Arg72-p53 neurons after ischemia. Furthermore, PC promoted neuroprotection against ischemia by controlling the p53/active caspase-3 pathway in Pro72-p53, but not in Arg72-p53 neurons. Finally, we found that good prognosis associated to TIA within 1 month prior to ischemic stroke was restricted to patients harboring the Pro72 allele. Our findings demonstrate that the Tp53 Arg72Pro SNP controls PC-promoted neuroprotection against a subsequent ischemic insult by modulating mitochondrial p53 stabilization and then modulates TIA-induced ischemic tolerance.


Subject(s)
Brain Ischemia/genetics , Cell Hypoxia/genetics , Ischemic Preconditioning/methods , Neurons/pathology , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/genetics , Aged , Aged, 80 and over , Animals , Apoptosis/genetics , Arginine/genetics , Brain Ischemia/prevention & control , Caspase 3/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cohort Studies , Electron Transport Complex IV/metabolism , Embryo, Mammalian , Excitatory Amino Acid Agonists/pharmacology , Female , Glucose/deficiency , Humans , Male , Membrane Potentials/genetics , Mice , Microtubule-Associated Proteins/metabolism , Middle Aged , N-Methylaspartate/pharmacology , Proline/genetics , Subcellular Fractions/metabolism , Subcellular Fractions/pathology
5.
Stroke ; 49(10): 2437-2444, 2018 10.
Article in English | MEDLINE | ID: mdl-30355102

ABSTRACT

Background and Purpose- The E3 ubiquitin ligase MDM2 (murine double minute 2) is the main negative regulator of the p53 protein-a key player in neuronal apoptosis after ischemia. A functional single-nucleotide polymorphism in the human MDM2 gene promoter (rs2279744) regulates MDM2 protein expression. We investigated whether the MDM2 SNP309, by controlling p53-mediated apoptosis, determines functional outcome after stroke. Methods- Primary cortical neurons were subjected to oxygen and glucose deprivation. Mice were subjected to ischemic (transient middle cerebral artery occlusion) or hemorrhagic (collagenase injection) stroke models. Protein and mRNA levels of MDM2 and p53 were measured in both neuronal and brain extracts. The interaction of MDM2 with p53 was disrupted by neuronal treatment with nutlin-3a. siRNA was used to knockdown MDM2 expression. We analyzed the link between the MDM2 SNP309 and functional outcome, measured by the modified Rankin Scale scores, in 2 independent hospital-based stroke cohorts: ischemic stroke cohort (408 patients) and intracerebral hemorrhage cohort (128 patients). Results- Experimental stroke and oxygen and glucose deprivation induced the expression of MDM2 in the brain and neurons, respectively. Moreover, oxygen and glucose deprivation promoted MDM2 binding with p53 in neurons. Disruption of the MDM2-p53 interaction with nutlin-3a, or MDM2 knockdown by siRNA, triggered p53 accumulation, which increased neuronal susceptibility to oxygen and glucose deprivation-induced apoptosis. Finally, we showed that patients harboring the G allele in the MDM2 promoter had higher MDM2 protein levels and showed better functional outcome after stroke than those harboring the T/T genotype. The T/T genotype was also associated with large infarct volume in ischemic stroke and increased lesion volume in patients with intracerebral hemorrhage. Conclusions- Our results reveal a novel role for the MDM2-p53 interaction in neuronal apoptosis after ischemia and show that the MDM2 SNP309 determines the functional outcome of patients after stroke.


Subject(s)
Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-mdm2/genetics , Recovery of Function/genetics , Stroke/genetics , Alleles , Animals , Genotype , Humans , Mice, Inbred C57BL , Polymorphism, Single Nucleotide/genetics , Risk Factors , Stroke/therapy
6.
Sci Rep ; 8(1): 1610, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371613

ABSTRACT

Brain preconditioning (PC) refers to a state of transient tolerance against a lethal insult that can be evoked by a prior mild event. It is thought that PC may induce different pathways responsible for neuroprotection, which may involve the attenuation of cell damage pathways, including the apoptotic cell death. In this context, p53 is a stress sensor that accumulates during brain ischemia leading to neuronal death. The murine double minute 2 gene (MDM2), a p53-specific E3 ubiquitin ligase, is the main cellular antagonist of p53, mediating its degradation by the proteasome. Here, we study the role of MDM2-p53 pathway on PC-induced neuroprotection both in cultured neurons (in vitro) and rat brain (in vivo). Our results show that PC increased neuronal MDM2 protein levels, which prevented ischemia-induced p53 stabilization and neuronal death. Indeed, PC attenuated ischemia-induced activation of the p53/PUMA/caspase-3 signaling pathway. Pharmacological inhibition of MDM2-p53 interaction in neurons abrogated PC-induced neuroprotection against ischemia. Finally, the relevance of the MDM2-p53 pathway was confirmed in rat brain using a PC model in vivo. These findings demonstrate the key role of the MDM2-p53 pathway in PC-induced neuroprotection against a subsequent ischemic insult and poses MDM2 as an essential target in ischemic tolerance.


Subject(s)
Brain/pathology , Ischemia/pathology , Ischemic Preconditioning , Neurons/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Cell Survival , Cells, Cultured , Disease Models, Animal , Mice , Rats
7.
Proc Natl Acad Sci U S A ; 114(17): 4513-4518, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28396402

ABSTRACT

Disruption of neuronal morphology contributes to the pathology of neurodegenerative disorders such as Alzheimer's disease (AD). However, the underlying molecular mechanisms are unknown. Here, we show that postnatal deletion of Cdh1, a cofactor of the anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase in neurons [Cdh1 conditional knockout (cKO)], disrupts dendrite arborization and causes dendritic spine and synapse loss in the cortex and hippocampus, concomitant with memory impairment and neurodegeneration, in adult mice. We found that the dendrite destabilizer Rho protein kinase 2 (Rock2), which accumulates in the brain of AD patients, is an APC/CCdh1 substrate in vivo and that Rock2 protein and activity increased in the cortex and hippocampus of Cdh1 cKO mice. In these animals, inhibition of Rock activity, using the clinically approved drug fasudil, prevented dendritic network disorganization, memory loss, and neurodegeneration. Thus, APC/CCdh1-mediated degradation of Rock2 maintains the dendritic network, memory formation, and neuronal survival, suggesting that pharmacological inhibition of aberrantly accumulated Rock2 may be a suitable therapeutic strategy against neurodegeneration.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Dendritic Cells/physiology , Gene Expression Regulation/physiology , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Cdh1 Proteins/genetics , Cell Survival , Memory/drug effects , Memory/physiology , Mice , Mice, Knockout , Neurons/physiology , Protein Kinase Inhibitors/pharmacology , Signal Transduction , rho-Associated Kinases/genetics
8.
Sci Rep ; 5: 18180, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26658992

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase that regulates cell cycle progression in proliferating cells. To enter the S-phase, APC/C must be inactivated by phosphorylation of its cofactor, Cdh1. In post-mitotic cells such as neurons APC/C-Cdh1 complex is highly active and responsible for the continuous degradation of mitotic cyclins. However, the specific molecular pathway that determines neuronal cell cycle blockade in post-mitotic neurons is unknown. Here, we show that activation of glutamatergic receptors in rat cortical primary neurons endogenously triggers cyclin-dependent kinase-5 (Cdk5)-mediated phosphorylation of Cdh1 leading to its cytoplasmic accumulation and disassembly from the APC3 core protein, causing APC/C inactivation. Conversely, pharmacological or genetic inhibition of Cdk5 promotes Cdh1 ubiquitination and proteasomal degradation. Furthermore, we show that Cdk5-mediated phosphorylation and inactivation of Cdh1 leads to p27 depletion, which switches on the cyclin D1-cyclin-dependent kinase-4 (Cdk4)-retinoblastoma protein (pRb) pathway to allow the S-phase entry of neurons. However, neurons do not proceed through the cell cycle and die by apoptosis. These results indicate that APC/C-Cdh1 actively suppresses an aberrant cell cycle entry and death of neurons, highlighting its critical function in neuroprotection.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cadherins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 5/metabolism , Neurons/metabolism , Retinoblastoma Protein/metabolism , S Phase , Signal Transduction , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Animals , Antigens, CD , Caspase 3/metabolism , Cell Cycle , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glutamic Acid , Mitosis , Phosphorylation , Protein Stability , Rats
9.
J Neurosci ; 35(25): 9287-301, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26109654

ABSTRACT

The survival of postmitotic neurons needs continuous degradation of cyclin B1, a mitotic protein accumulated aberrantly in the damaged brain areas of Alzheimer's disease and stroked patients. Degradation of cyclin B1 takes place in the proteasome after ubiquitylation by the anaphase-promoting complex/cyclosome (APC/C)-cadherin 1 (Cdh1), an E3 ubiquitin ligase that is highly active in neurons. However, during excitotoxic damage-a hallmark of neurological disorders-APC/C-Cdh1 is inactivated, causing cyclin B1 stabilization and neuronal death through an unknown mechanism. Here, we show that an excitotoxic stimulus in rat cortical neurons in primary culture promotes cyclin B1 accumulation in the mitochondria, in which it binds to, and activates, cyclin-dependent kinase-1 (Cdk1). The cyclin B1-Cdk1 complex in the mitochondria phosphorylates the anti-apoptotic protein B-cell lymphoma extra-large (Bcl-xL), leading to its dissociation from the ß subunit of F1Fo-ATP synthase. The subsequent inhibition of ATP synthase activity causes complex I oxidative damage, mitochondrial inner membrane depolarization, and apoptotic neuronal death. These results unveil a previously unrecognized role for mitochondrial cyclin B1 in the oxidative damage associated with neurological disorders.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis/physiology , Cyclin B1/metabolism , Cyclin-Dependent Kinases/metabolism , Neurons/metabolism , bcl-X Protein/metabolism , Animals , Blotting, Western , CDC2 Protein Kinase , Cell Survival , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Immunoprecipitation , Mitochondria/metabolism , Mutagenesis, Site-Directed , Nerve Degeneration/metabolism , Oxidative Stress/physiology , Protein Binding , RNA, Small Interfering , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
Free Radic Biol Med ; 75 Suppl 1: S4-5, 2014 Oct.
Article in English | MEDLINE | ID: mdl-26461363

ABSTRACT

The morphology of the adult brain is the result of a delicate balance between the symmetric divisions to maintain the progenitor cell pool, and the asymmetric divisions to generate a newly differentiated neuron. Neurogenesis is a complex process that relies on an as yet unknown molecular switch that tightly coordinates the cell cycle exit with the start of the differentiation process. The cell cycle length is a key factor that determines the balance between the maintenance of progenitor cells and neuronal differentiation. In fact, neurogenesis in the cerebral cortex is stimulated by lengthening the G1 phase and delayed by shortening it. The anaphase-promoting complex/cyclosome (APC/C) cofactor, Cdh1, regulates mitosis exit and G1-phase length in proliferating cells. Here we assessed whether APC/C-Cdh1 activity would be responsible for the switch from progenitor cells cycling to neurogenesis in the cerebral cortex. We use an embryo-restricted Cdh1 knockout mouse model and show that functional APC/C-Cdh1 ubiquitin ligase activity is required for both terminal differentiation of cortical neurons in vitro and neurogenesis in vivo. Further, genetic ablation of Cdh1 impairs the ability of APC/C to promote neurogenesis by delaying the exit of the progenitor cells from the cell cycle. This causes replicative stress and p53-mediated apoptotic death resulting in decreased number of cortical neurons and cortex size. These results demonstrate that APC/C-Cdh1 coordinates cortical neurogenesis and size, thus posing Cdh1 in the molecular pathogenesis of congenital neurodevelopmental disorders, such as microcephaly.

11.
Nat Commun ; 4: 2879, 2013.
Article in English | MEDLINE | ID: mdl-24301314

ABSTRACT

The morphology of the adult brain is the result of a delicate balance between neural progenitor proliferation and the initiation of neurogenesis in the embryonic period. Here we assessed whether the anaphase-promoting complex/cyclosome (APC/C) cofactor, Cdh1--which regulates mitosis exit and G1-phase length in dividing cells--regulates neurogenesis in vivo. We use an embryo-restricted Cdh1 knockout mouse model and show that functional APC/C-Cdh1 ubiquitin ligase activity is required for both terminal differentiation of cortical neurons in vitro and neurogenesis in vivo. Further, genetic ablation of Cdh1 impairs the ability of APC/C to promote neurogenesis by delaying the exit of the progenitor cells from the cell cycle. This causes replicative stress and p53-mediated apoptotic death resulting in decreased number of cortical neurons and cortex size. These results demonstrate that APC/C-Cdh1 coordinates cortical neurogenesis and size, thus posing Cdh1 in the molecular pathogenesis of congenital neurodevelopmental disorders, such as microcephaly.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Cerebral Cortex/embryology , Neurogenesis , Neurons/cytology , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Apoptosis , Cdh1 Proteins/genetics , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Female , Male , Mice , Mice, Knockout , Neurons/enzymology , Neurons/metabolism , Organ Size
12.
J Exp Med ; 208(3): 429-37, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21357744

ABSTRACT

The functional outcome after stroke is unpredictable; it is not accurately predicted by clinical pictures upon hospital admission. The presence of apoptotic neurons in the ischemic penumbra and perihematoma area may account for poor prognosis, but whether the highly variable stroke outcome reflects differences in genetic susceptibility to apoptosis is elusive. The p53 tumor suppressor protein, an important transcriptional regulator of apoptosis, naturally occurs in humans in two variants with single nucleotide polymorphisms resulting in Arg or Pro at residue 72. We show that poor functional outcome after either ischemic or hemorrhagic stroke was linked to the Arg/Arg genotype. This genotype was also associated with early neurological deterioration in ischemic stroke and with increased residual cavity volume in intracerebral hemorrhage. In primary cultured neurons, Arg(72)-p53, but not Pro(72)-p53, interacted directly with mitochondrial Bcl-xL and activated the intrinsic apoptotic pathway, increasing vulnerability to ischemia-induced apoptotic cell death. These results suggest that the Tp53 Arg/Arg genotype governs neuronal vulnerability to apoptosis and can be considered as a genetic marker predicting poor functional outcome after stroke.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Stroke/genetics , Tumor Suppressor Protein p53/genetics , Aged , Apoptosis/genetics , Apoptosis/physiology , Brain Ischemia/genetics , Brain Ischemia/physiopathology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/physiopathology , Female , Genetic Association Studies , Genetic Markers/genetics , Genetic Markers/physiology , Genotype , Humans , Male , Polymorphism, Single Nucleotide/physiology , Prognosis , Stroke/diagnosis , Tumor Suppressor Protein p53/physiology
13.
J Neurochem ; 113(4): 819-25, 2010 May.
Article in English | MEDLINE | ID: mdl-20180881

ABSTRACT

Neuroblastoma is a sympathetic nervous system tumour whose degree of malignancy, prognosis and therapy resistance has been associated with the amplification of MYCN oncogene. However, the molecular pathway responsible for such resistance is unknown. To contribute addressing this issue, in this study, we have compared the vulnerability of four human neuroblastoma cell lines differentially amplifying MYCN, namely SK-N-BE-2 and IMR-32 (MYCN-amplified cells) and SH-SY5Y and SK-N-SH (MCYN-non-amplified cells), to H(2)O(2)-mediated apoptotic death. We found that the high resistance of the MYCN-amplified neuroblastoma cells against oxidative damage can be accounted for by their greater expression of both the mRNA and protein of the catalytic subunit of glutamate-cysteine ligase (GCL(cat)), the rate-limiting step in GSH biosynthesis. Furthermore, we found that MYCN directly binds to an E-box containing GCL(cat) promoter and that over-expression of MYCN in MYCN-non-amplified cells stimulated GCL(cat) expression and provided resistance to oxidative damage; whereas knock down of MYCN in MYCN-amplified cells decreased GCL(cat) expression and sensitized them to oxidative damage. Finally, GCL(cat) knock down enhanced the vulnerability of MYCN-amplified cells to oxidative damage. These results demonstrate that regulation of GCL(cat) by MYCN accounts for the survival of neuroblastoma cells against oxidative damage, and suggest that GCL should be considered a potential therapeutic target for the treatment of MYCN-amplified neuroblastoma.


Subject(s)
Drug Resistance, Neoplasm/genetics , Glutamate-Cysteine Ligase/genetics , Neuroblastoma/enzymology , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Oxidative Stress/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Down-Regulation/genetics , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/chemistry , Humans , Hydrogen Peroxide/toxicity , N-Myc Proto-Oncogene Protein , Neuroblastoma/drug therapy , Oxidants/toxicity , Oxidative Stress/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Transcriptional Activation/physiology , Up-Regulation/physiology
14.
J Neurochem ; 112(1): 56-65, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19818102

ABSTRACT

High levels of unconjugated bilirubin (UCB) may initiate encephalopathy in neonatal life, mainly in pre-mature infants. The molecular mechanisms of this bilirubin-induced neurologic dysfunction (BIND) are not yet clarified and no neuroprotective strategy is currently worldwide accepted. Here, we show that UCB, at conditions mimicking those of hyperbilirubinemic newborns (50 microM UCB in the presence of 100 muM human serum albumin), rapidly (within 1 h) inhibited cytochrome c oxidase activity and ascorbate-driven oxygen consumption in 3 days in vitro rat cortical neurons. This was accompanied by a bioenergetic and oxidative crisis, and apoptotic cell death, as judged by the collapse of the inner-mitochondrial membrane potential, increased glycolytic activity, superoxide anion radical production, and ATP release, as well as disruption of glutathione redox status. Furthermore, the antioxidant compound glycoursodeoxycholic acid (GUDCA) fully abrogated UCB-induced cytochrome c oxidase inhibition and significantly prevented oxidative stress, metabolic alterations, and cell demise. These results suggest that the neurotoxicity associated with neonatal bilirubin-induced encephalopathy occur through a dysregulation of energy metabolism, and supports the notion that GUDCA may be useful in the treatment of BIND.


Subject(s)
Apoptosis/physiology , Bilirubin/toxicity , Cell Differentiation/physiology , Cerebral Cortex/enzymology , Electron Transport Complex IV/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Ursodeoxycholic Acid/analogs & derivatives , Animals , Apoptosis/drug effects , Bilirubin/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Electron Transport Complex IV/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/toxicity , Female , Humans , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Pregnancy , Rats , Rats, Wistar , Ursodeoxycholic Acid/physiology
15.
EMBO J ; 27(20): 2736-45, 2008 Oct 22.
Article in English | MEDLINE | ID: mdl-18818692

ABSTRACT

Anaphase-promoting complex/cyclosome (APC/C), an E3 ubiquitin ligase that destabilizes cell cycle proteins, is activated by Cdh1 in post-mitotic neurons, where it regulates axonal growth, synaptic plasticity and survival. The APC/C-Cdh1 substrate, cyclin B1, has been found to accumulate in degenerating brain areas in Alzheimer's disease and stroke. This highlights the importance of elucidating cyclin B1 regulation by APC/C-Cdh1 in neurons under stress conditions relevant to neurological disease. Here, we report that stimulation of N-methyl-D-aspartate receptors (NMDARs) that occurs in neurodegenerative diseases promoted the accumulation of cyclin B1 in the nuclei of cortical neurons; this led the neurons to undergo apoptotic death. Moreover, we found that the Ser-40, Thr-121 and Ser-163 triple phosphorylation of Cdh1 by the cyclin-dependent kinase-5 (Cdk5)-p25 complex was necessary and sufficient for cyclin B1 stabilization and apoptotic death after NMDAR stimulation. These results reveal Cdh1 as a novel Cdk5 substrate that mediates cyclin B1 neuronal accumulation in excitotoxicity.


Subject(s)
Cadherins/metabolism , Cyclin B/metabolism , Cyclin-Dependent Kinase 5/metabolism , Animals , Apoptosis , Cell Nucleus/metabolism , Cells, Cultured , Cyclin B1 , Models, Biological , Neurons/metabolism , Phosphorylation , Plasmids/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/chemistry
16.
Biochim Biophys Acta ; 1777(7-8): 789-93, 2008.
Article in English | MEDLINE | ID: mdl-18455501

ABSTRACT

Besides its essential role at regulating neural functions through cyclic GMP, nitric oxide is emerging as an endogenous physiological modulator of energy conservation for the brain. Thus, nitric oxide inhibits cytochrome c oxidase activity in neurones and glia, resulting in down-regulation of mitochondrial energy production. The subsequent increase in AMP facilitates the activation of 5'-AMP-dependent protein kinase, which rapidly triggers the activation of 6-phosphofructo-1-kinase--the master regulator of the glycolytic pathway--and Glut1 and Glut3--the main glucose transporters in the brain. In addition, nitric oxide activates glucose-6-phosphate dehydrogenase, the first and rate-limiting step of the pentose-phosphate pathway. Here, we review recent evidences suggesting that nitric oxide exerts a fine control of neuronal energy metabolism by tuning the balance of glucose-6-phosphate consumption between glycolysis and pentose-phosphate pathway. This may have important implications for our understanding of the mechanisms controlling neuronal survival during oxidative stress and bioenergetic crisis.


Subject(s)
Cell Survival/drug effects , Glycolysis , Neurons/physiology , Nitric Oxide/pharmacology , Pentose Phosphate Pathway/physiology , Animals , Energy Metabolism , Glycolysis/drug effects , Homeostasis , Humans , Neurons/cytology , Neurons/drug effects , Nitric Oxide/physiology , Nitric Oxide Synthase/metabolism , Pentose Phosphate Pathway/drug effects , Peroxynitrous Acid/pharmacology , Phosphofructokinase-1/metabolism
17.
J Mol Neurosci ; 34(1): 77-87, 2008.
Article in English | MEDLINE | ID: mdl-18040888

ABSTRACT

Oxidative damage has been reported to be involved in the pathogenesis of diabetic neuropathy and neurodegenerative diseases. Recent evidence suggests that the antidiabetic drug metformin prevents oxidative stress-related cellular death in non-neuronal cell lines. In this report, we point to the direct neuroprotective effect of metformin, using the etoposide-induced cell death model. The exposure of intact primary neurons to this cytotoxic insult induced permeability transition pore (PTP) opening, the dissipation of mitochondrial membrane potential (DeltaPsim), cytochrome c release, and subsequent death. More importantly, metformin, together with the PTP classical inhibitor cyclosporin A (CsA), strongly mitigated the activation of this apoptotic cascade. Furthermore, the general antioxidant N-acetyl-L: -cysteine also prevented etoposide-promoted neuronal death. In addition, metformin was shown to delay CsA-sensitive PTP opening in permeabilized neurons, as triggered by a calcium overload, probably through its mild inhibitory effect on the respiratory chain complex I. We conclude that (1) etoposide-induced neuronal death is partly attributable to PTP opening and the disruption of DeltaPsim, in association with the emergence of oxidative stress, and (2) metformin inhibits this PTP opening-driven commitment to death. We thus propose that metformin, beyond its antihyperglycemic role, can also function as a new therapeutic tool for diabetes-associated neurodegenerative disorders.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/drug effects , Metformin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cyclosporine/pharmacology , Cytochromes c/metabolism , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/physiopathology , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Metformin/therapeutic use , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Oxidative Stress/physiology , Rats , Rats, Wistar
18.
J Neurochem ; 102(1): 194-205, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17302912

ABSTRACT

Peroxynitrite is usually considered as a neurotoxic nitric oxide-derivative. However, an increasing body of evidence suggests that, at low concentrations, peroxynitrite affords transient cytoprotection, both in vitro and in vivo. Here, we addressed the signaling mechanism responsible for this effect, and found that rat cortical neurons in primary culture acutely exposed to peroxynitrite (0.1 mmol/L) rapidly elicited Akt-Ser(473) phosphorylation. Inhibition of phosphoinositide-3-kinase (PI3K)/Akt pathway with wortmannin or Akt small hairpin RNA (shRNA) abolished the ability of peroxynitrite to prevent etoposide-induced apoptotic death. Endogenous peroxynitrite formation by short-term incubation of neurons with glutamate stimulated Akt-Ser(473) phosphorylation, whereas Akt shRNA enhanced the vulnerability of neurons against glutamate. We further show that Akt-Ser(473) phosphorylation was consequence of the oxidizing, but not the nitrating properties of peroxynitrite. Peroxynitrite failed to nitrate or phosphorylate neurotrophin tyrosine kinase receptors (Trks), and it did not modify the ability of brain-derived neurotrophic factor (BDNF), to phosphorylate its cognate receptor, TrkB; however, peroxynitrite enhanced BDNF-mediated Akt-Ser(473) phosphorylation. Finally, we found that peroxynitrite-stimulated Akt-Ser(473) phosphorylation was associated with an increased proportion of oxidized phosphoinositide phosphatase, PTEN, in neurons. Moreover, peroxynitrite prevented the increase of apoptotic neuronal death caused by over-expression of PTEN. Thus, peroxynitrite exerts neuroprotection by inhibiting PTEN, hence activating the anti-apoptotic PI3K/Akt pathway in primary neurons.


Subject(s)
Neuroprotective Agents/pharmacology , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Peroxynitrous Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Etoposide/antagonists & inhibitors , Etoposide/pharmacology , Flow Cytometry , Humans , Immunoprecipitation , NADP/metabolism , Nerve Degeneration/prevention & control , Oxidation-Reduction , Peroxynitrous Acid/chemical synthesis , Phosphorylation , Plasmids/genetics , RNA/biosynthesis , RNA/genetics , Transfection
19.
J Biol Chem ; 280(47): 38992-9001, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16183645

ABSTRACT

Glutathione deficiency is an early biochemical feature that occurs during apoptotic neuronal death associated with certain neurological disorders such as Parkinson disease. However, whether specific targeting of glutathione biosynthesis in neurons is sufficient to trigger neurodegeneration remains undetermined. To address this issue, we used a vector-based small hairpin RNA (shRNA) strategy to knock down each subunit of glutamate-cysteine ligase (GCL; gamma-glutamylcysteine synthetase), the heterodimeric enzyme that catalyzes the rate-limiting step of glutathione biosynthesis. Independent targeting of the catalytic and modulatory subunits by shRNA caused disruption of GCL as assessed by Northern and Western blotting, enzyme activity, and glutathione concentrations. Silencing each subunit in primary cortical neurons spontaneously elicited time-dependent apoptotic death, an effect that was synergistic with glutamate or nitric oxide treatment. Moreover, neuronal apoptosis by GCL knockdown was rescued by expressing the corresponding subunit full-length cDNA carrying silent mutations within the shRNA target cDNA sequence and by incubating neurons with gamma-glutamylcysteine or glutathione ethyl ester. In contrast, supplying glutathione precursors to neurons from co-cultured astrocytes did not prevent the apoptotic death triggered by GCL knockdown. Finally, overexpressing the catalytic (but not modulatory) GCL subunit full-length cDNA increased enzyme activity and glutathione concentrations, yielding neurons more resistant to glutamate- or nitric oxide-mediated apoptosis. Thus, specific and independent disruption of each subunit of GCL in neurons can be said to cause a primary decrease in glutathione that is sufficient to promote neurodegeneration.


Subject(s)
Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/chemistry , Neurons/cytology , Neurons/enzymology , RNA, Small Interfering/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Survival , Cells, Cultured , Glutamate-Cysteine Ligase/genetics , Glutamic Acid/toxicity , Humans , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Neurons/drug effects , Nitric Oxide/toxicity , Protein Subunits , RNA Interference , Rats
20.
J Neurosci Res ; 79(1-2): 166-71, 2005.
Article in English | MEDLINE | ID: mdl-15573411

ABSTRACT

There is an increasing body of evidence demonstrating that inhibition of cytochrome c oxidase by nitric oxide (NO) may be one more step in a signaling cascade involved in the physiologic regulation of cell functions. For example, in both astrocytes and neurons the inhibition of mitochondrial respiration by endogenously produced NO induces transient and modest decreases in cellular ATP concentrations. This mitochondrial impairment may serve as a cellular sensor of energy charges, hence modulating metabolic pathways, such as glycolysis, through AMP-activated protein kinase (AMPK) in astrocytes. In neurons, the NO derivative peroxynitrite anion triggers signaling pathways leading to glucose oxidation through the pentose-phosphate pathway to form reducing equivalents in the form of NADPH. The modulation of these metabolic pathways by nitric oxide or its derivatives may be important for understanding the mechanisms by which this free radical affects neuronal death or survival.


Subject(s)
Glucose/metabolism , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide/physiology , Respiration/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Brain/cytology , Homeostasis/physiology , Humans , Mitochondria/metabolism , Models, Biological , Nitric Oxide/biosynthesis , Peroxynitrous Acid/pharmacology , Peroxynitrous Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...