Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 22(6): 874-883, 2017 06.
Article in English | MEDLINE | ID: mdl-27113998

ABSTRACT

A recent genome-wide association meta-analysis for Alzheimer's disease (AD) identified 19 risk loci (in addition to APOE) in which the functional genes are unknown. Using Drosophila, we screened 296 constructs targeting orthologs of 54 candidate risk genes within these loci for their ability to modify Tau neurotoxicity by quantifying the size of >6000 eyes. Besides Drosophila Amph (ortholog of BIN1), which we previously implicated in Tau pathology, we identified p130CAS (CASS4), Eph (EPHA1), Fak (PTK2B) and Rab3-GEF (MADD) as Tau toxicity modulators. Of these, the focal adhesion kinase Fak behaved as a strong Tau toxicity suppressor in both the eye and an independent focal adhesion-related wing blister assay. Accordingly, the human Tau and PTK2B proteins biochemically interacted in vitro and PTK2B co-localized with hyperphosphorylated and oligomeric Tau in progressive pathological stages in the brains of AD patients and transgenic Tau mice. These data indicate that PTK2B acts as an early marker and in vivo modulator of Tau toxicity.


Subject(s)
Focal Adhesion Kinase 2/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Animals , Biomarkers , Disease Models, Animal , Drosophila/genetics , Focal Adhesion Kinase 2/metabolism , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Risk Factors , tau Proteins/genetics
2.
Neurobiol Learn Mem ; 125: 274-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26463507

ABSTRACT

The benefits of neuromodulatory procedures as a possible therapeutic application for cognitive rehabilitation have increased with the progress made in non-invasive modes of brain stimulation in aged-related disorders. Transcranial magnetic stimulation (TMS) is a non-invasive method used to examine multiple facets of the human brain and to ameliorate the impairment in cognition caused by Alzheimer's disease (AD). The present study was designed to evaluate how a chronic TMS treatment could improve learning and memory functions after sleep deprivation (SD) in old Octodon degus. SD was executed by gently handling to keep the animals awake throughout the night. Thirty young and twenty-four old O. degus females were divided in six groups (control, acute and chronic TMS treatment). Behavioral tests included; Radial Arm Maze (RAM), Barnes Maze (BM) and Novel Object Recognition (NOR). Although learning and memory functions improved in young animals with only one session of TMS treatment, a significant improvement in cognitive performance was seen in old animals after 4 and 7days of TMS, depending on the task that was performed. No side effects were observed following, which showed therapeutic potential for improving age-related cognitive performance.


Subject(s)
Aging/physiology , Brain/physiopathology , Cognition/physiology , Memory/physiology , Sleep Deprivation/physiopathology , Spatial Learning/physiology , Animals , Behavior, Animal/physiology , Female , Octodon , Transcranial Magnetic Stimulation
3.
Neurotox Res ; 28(4): 361-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26194615

ABSTRACT

Sleep is indispensable for maintaining regular daily life activities and is of fundamental physiological importance for cognitive performance. Sleep deprivation (SD) may affect learning capacity and the ability to form new memories, particularly with regard to hippocampus-dependent tasks. Transcranial magnetic stimulation (TMS) is a non-invasive procedure of electromagnetic induction that generates electric currents, activating nearby nerve cells in the stimulated cortical area. Several studies have looked into the potential therapeutic use of TMS. The present study was designed to evaluate how TMS could improve learning and memory functions following SD in Octodon degus. Thirty juvenile (18 months old) females were divided into three groups (control, acute, and chronic TMS treatment-with and without SD). TMS-treated groups were placed in plastic cylindrical cages designed to keep them immobile, while receiving head magnetic stimulation. SD was achieved by gently handling the animals to keep them awake during the night. Behavioral tests included radial arm maze (RAM), Barnes maze (BM), and novel object recognition. When TMS treatment was applied over several days, there was significant improvement of cognitive performance after SD, with no side effects. A single TMS session reduced the number of errors for the RAM test and improved latency and reduced errors for the BM test, which both evaluate spatial memory. Moreover, chronic TMS treatment brings about a significant improvement in both spatial and working memories.


Subject(s)
Cognition Disorders/physiopathology , Learning/physiology , Memory/physiology , Sleep Deprivation/complications , Transcranial Magnetic Stimulation , Animals , Brain/physiopathology , Cognition Disorders/etiology , Female , Octodon , Recognition, Psychology/physiology
4.
Biochim Biophys Acta ; 1842(4): 654-64, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440524

ABSTRACT

The splicing of the microtubule-associated protein Tau is regulated during development and is found to be deregulated in a growing number of pathological conditions such as myotonic dystrophy type I (DM1), in which a reduced number of isoforms is expressed in the adult brain. DM1 is caused by a dynamic and unstable CTG repeat expansion in the DMPK gene, resulting in an RNA bearing long CUG repeats (n>50) that accumulates in nuclear foci and sequesters CUG-binding splicing factors of the muscle blind-like (MBNL) family, involved in the splicing of Tau pre-mRNA among others. However, the precise mechanism leading to Tau mis-splicing and the role of MBNL splicing factors in this process are poorly understood. We therefore used new Tau minigenes that we developed for this purpose to determine how MBNL1 and MBNL2 interact to regulate Tau exon 2 splicing. We demonstrate that an intronic region 250 nucleotides downstream of Tau exon 2 contains cis-regulatory splicing enhancers that are sensitive to MBNL and that bind directly to MBNL1. Both MBNL1 and MBNL2 act as enhancers of Tau exon 2 inclusion. Intriguingly, the interaction of MBNL1 and MBNL2 is required to fully reverse the mis-splicing of Tau exon 2 induced by the trans-dominant effect of long CUG repeats, similar to the DM1 condition. In conclusion, both MBNL1 and MBNL2 are involved in the regulation of Tau exon 2 splicing and the mis-splicing of Tau in DM1 is due to the combined inactivation of both.


Subject(s)
Exons , Myotonic Dystrophy/genetics , RNA-Binding Proteins/physiology , Response Elements , tau Proteins/genetics , Base Sequence , Cell Line, Tumor , Humans , Molecular Sequence Data , RNA Splicing
5.
Curr Alzheimer Res ; 8(6): 633-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21605043

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder histologically defined by the cerebral accumulation of amyloid deposits and neurofibrillary tangles composed of hyperphosphorylated tau proteins. Loss of basal forebrain cholinergic neurons is another hallmark of the disease thought to contribute to the cognitive dysfunctions. To this date, the mechanisms underlying cholinergic neurons degeneration remain uncertain. The present study aimed to investigate the relationship between neurofibrillary degeneration and cholinergic defects in AD using THY-Tau22 transgenic mouse model exhibiting a major hippocampal AD-like tau pathology and hyperphosphorylated tau species in the septohippocampal pathway. Here, we report that at a time THY-Tau22 mice display strong reference memory alterations, the retrograde transport of fluorogold through the septohippocampal pathway is altered. This impairment is associated with a significant reduction in the number of choline acetyltransferase (ChAT)-immunopositive cholinergic neurons in the medial septum. Analysis of nerve growth factor (NGF) levels supports an accumulation of the mature neurotrophin in the hippocampus of THY-Tau22 mice, consistent with a decrease of its uptake or retrograde transport by cholinergic terminals. Finally, our data strongly support that tau pathology could be instrumental in the cholinergic neuronal loss observed in AD.


Subject(s)
Brain/pathology , Cholinergic Neurons/pathology , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Animals , Brain/metabolism , Cholinergic Neurons/metabolism , Maze Learning/physiology , Memory/physiology , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurons/metabolism , Neurons/pathology , tau Proteins/genetics
6.
Rev Neurol ; 47(5): 253-60, 2008.
Article in Spanish | MEDLINE | ID: mdl-18780272

ABSTRACT

INTRODUCTION: Stroke leads the list of causes of disability in adults and represents the second leading cause of death worldwide. Knowledge about the pathophysiology of ischemic stroke has improved substantially over the past 25 years, and, as a result of this, new therapeutic strategies have been developed with two main aims: restoration of cerebral flow and the minimization of the deleterious effects of ischemia on neurons. Although so far there are no drugs approved for the neuroprotection therapy in stroke, there are some compounds with promising results. DEVELOPMENT: This paper makes a critical review of several studies on the preclinical stroke neuroprotection with drugs aimed to protect the brain tissue adjacent to the damaged central area or ischemic penumbra zone until either the physiological mechanisms or the treatment stop the ischemic insult. We expose the potential neuroprotective properties of these treatments mainly based on inhibiting excitotoxicity processes mediated by gamma-aminobutyric acid receptors, glutamate release and interacting with ion channels such as calcium and sodium. We focus on drugs which have shown to be capable of modulating intracellular degenerative pathways in mitochondria mediated apoptosis or the expression of apoptotic proteins in experimental models. CONCLUSION: It is very likely that the neuroprotective effects require a poly-drug therapy that combines different mechanisms of action.


Subject(s)
Neuroprotective Agents , Stroke/drug therapy , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Drug Therapy, Combination , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Stroke/pathology , Stroke/physiopathology , Thrombolytic Therapy
7.
Rev Neurol ; 47(1): 31-8, 2008.
Article in Spanish | MEDLINE | ID: mdl-18592478

ABSTRACT

INTRODUCTION: During the last decade, the neuroprotective effects of minocycline have been a matter of an intense debate. A broad amount of contradictory studies can be found in the scientific literature, going from neuroprotection to the exacerbation of toxicity in diverse experimental models. Such differences could be the result of minocycline acting on multiple pharmacological targets. DEVELOPMENT: In the present review we will go over these pharmacological targets and the effects derived from their modulation by minocycline. Among others, its antioxidant activity derived from its chemical structure or its modulator effect on several enzymes such as nitric oxide synthase will be reviewed. Furthermore, the effects of minocycline on the intracellular pathways implicated in neurodegenerative processes including apoptosis stages, activation decision and execution will be addressed. CONCLUSIONS: All the mechanisms described herein have not escaped to a scientific community needed of new therapeutic drugs for the treatment of neurodegenerative conditions. However, the sparse clinical trials carried out so far are mainly aimed at assessing its tolerability and safety or are still in progress. We believe that more studies, both clinical and pre-clinical, should be carried out in order to ascertain the therapeutic window and the neurodegenerative disorders in which minocycline could be useful.


Subject(s)
Minocycline/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Apoptosis/drug effects , Humans , Minocycline/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology
8.
Curr Drug Deliv ; 4(3): 225-31, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17627496

ABSTRACT

In this review we explore and integrate the knowledge of the plausible pharmacological targets that could explain the new application for the well known semi-synthetic, tetracycline-derivate minocycline as a cytoprotective drug. In doing so, we will analyze the possible mechanisms to elucidate the potential cytoprotective properties of minocycline. We address its anti-oxidant action ranging from its structure to its capacity to modulate the expression of oxidant-related enzymes such as nitric oxide synthase. The pharmacological targets responsible for its anti-inflammatory effects are surveyed. The effects of this antibiotic are making its marks on intracellular pathways related to neurodegenerative processes such as mitochondrially-mediated apoptosis, including minocycline-modulated effects on the expression of apoptotic proteins. Finally, we will explore the effects of minocycline on metalloproteinases, enzymes implicated in the modulation of cerebrovascular post-ischemic oxidative reperfusion injury, and new targets. In conclusion, we shed new light on the shadowy controversy of minocycline's potential cytoprotective mechanisms and targets of action.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Cytoprotection , Minocycline/pharmacology , Apoptosis/drug effects , Drug Delivery Systems , Humans , Metalloproteases/drug effects , Mitochondria/drug effects , Neurodegenerative Diseases/drug therapy , Nitric Oxide Synthase/drug effects
9.
Mol Pharmacol ; 71(3): 736-43, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17172466

ABSTRACT

Malonate, an inhibitor of mitochondrial complex II, is a widely used toxin to study neurodegeneration in Huntington's disease and ischemic stroke. We have shown previously that malonate increased reactive oxygen species (ROS) production in human SH-SY5Y neuroblastoma cells, leading to oxidative stress, cytochrome c release, and apoptotic cell death. Expression of a green fluorescent protein-Bax fusion protein in SH-SY5Y neuroblastoma cells demonstrated a Bax redistribution from the cytosol to mitochondria after 12 to 24 h of malonate treatment that coincided with mitochondrial potential collapse and chromatin condensation. Inhibition of Bax translocation using furosemide, as well as Bax gene deletion, afforded significant protection against malonate-induced apoptosis. Further experiments revealed that malonate induced a prominent increase in the level of activated p38 mitogen-activated protein (MAP) kinase and that treatment with the p38 MAP kinase inhibitor SKF86002 potently blocked malonate-induced Bax translocation and apoptosis. Treatment with vitamin E diminished ROS production, reduced the activation status of p38 MAP kinase, inhibited Bax translocation, and protected against malonate-induced apoptosis. Our data suggest that malonate-induced ROS production and subsequent p38 MAP kinase activation mediates the activation of the pro-apoptotic Bax protein to induce mitochondrial membrane permeabilization and neuronal apoptosis.


Subject(s)
Apoptosis/drug effects , Cytochromes c/metabolism , Malonates/pharmacology , Mitochondria/drug effects , Reactive Oxygen Species , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured , Malondialdehyde/analysis , Mitochondria/metabolism , Protein Transport/drug effects , Rats
10.
Neurobiol Dis ; 24(2): 296-307, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16978869

ABSTRACT

Parkinson disease (PD) is the second-most common age-related neurodegenerative disease and is characterized by the selective destruction of dopaminergic neurons. Increasing evidence indicates that oxidative stress plays a crucial role in the pathogenesis of idiopathic PD. Anti-oxidant agents including catalase, manganese porphyrin and pyruvate confer cytoprotection to different cell cultures when challenged with 6-hydroxydopamine (6-OHDA). Herein we used rat cerebellar granular cell cultures to ascertain the plausible cellular pathways involved in pyruvate-induced cytoprotection against 0.1 mM 6-OHDA. Pyruvate provided cytoprotection in a concentration-dependent manner (2-10 mM). Consistent with its well-established anti-oxidant capacity, pyruvate (10 mM) prevented 6-OHDA-induced lipid peroxidation by blocking the rise in intracellular peroxides and maintaining the intracellular reduced glutathione (GSH) levels. Further experiments revealed that pyruvate increased Akt, but not extracellular signal-regulated kinase phosphorylation. Moreover, phosphatidylinositol 3-kinase (PI3K) inhibitors attenuated pyruvate-induced cytoprotection indicating that PI3K-mediated Akt activation is necessary for pyruvate to induce cytoprotection. On the other hand, pyruvate also up-regulated glutathione peroxidase mRNA levels, but not those of the anti-oxidant enzymes superoxide dismutase-1 and -2, catalase or the anti-apoptotic oncogenes Bcl-2 or Bcl-xL. In summary, our results strongly suggest that pyruvate, besides the anti-oxidant properties related to its structure, exerts cytoprotective actions by activating different anti-apoptotic routes that include gene regulation and Akt pathway activation.


Subject(s)
Cerebellar Cortex/drug effects , Nerve Degeneration/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Pyruvic Acid/pharmacology , Animals , Animals, Newborn , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Cerebellar Cortex/metabolism , Cerebellar Cortex/physiopathology , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyruvic Acid/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
11.
Neurobiol Dis ; 20(2): 384-91, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16242643

ABSTRACT

Experimental and clinical studies support the view that the semisynthetic tetracycline minocycline exhibits neuroprotective roles in several models of neurodegenerative diseases, including ischemia, Huntington, Parkinson diseases, and amyotrophic lateral sclerosis. However, recent evidence indicates that minocycline does not always present beneficial actions. For instance, in an in vivo model of Huntington's disease, it fails to afford protection after malonate intrastriatal injection. Moreover, it reverses the neuroprotective effect of creatine in nigrostriatal dopaminergic neurons. This apparent contradiction prompted us to analyze the effect of this antibiotic on malonate-induced cell death. We show that, in rat cerebellar granular cells, the succinate dehydrogenase inhibitor malonate induces cell death in a concentration-dependent manner. By using DFCA, monochlorobimane and 10-N-nonyl-Acridin Orange to measure, respectively, H2O2-derived oxidant species and reduced forms of GSH and cardiolipin, we observed that malonate induced reactive oxygen species (ROS) production to an extent that surpasses the antioxidant defense capacity of the cells, resulting in GSH depletion and cardiolipin oxidation. The pre-treatment for 4 h with minocycline (10-100 microM) did not present cytoprotective actions. Moreover, minocycline failed to block ROS production and to abrogate malonate-induced oxidation of GSH and cardiolipin. Additional experiments revealed that minocycline was also unsuccessful to prevent the mitochondrial swelling induced by malonate. Furthermore, malonate did not induce the expression of the iNOS, caspase-3, -8, and -9 genes which have been shown to be up-regulated in several models where minocycline resulted cytoprotective. In addition, malonate-induced down-regulation of the antiapoptotic gene Bcl-2 was not prevented by minocycline, controversially the mechanism previously proposed to explain minocycline protective action. These results suggest that the minocycline protection observed in several neurodegenerative disease models is selective, since it is absent from cultured cerebellar granular cells challenged with malonate.


Subject(s)
Apoptosis/drug effects , Malonates/antagonists & inhibitors , Minocycline/pharmacology , Nerve Degeneration/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Apoptosis/physiology , Cardiolipins/drug effects , Cardiolipins/metabolism , Caspases/drug effects , Caspases/metabolism , Cells, Cultured , Cerebellar Cortex/drug effects , Cerebellar Cortex/metabolism , Cerebellar Cortex/pathology , Dose-Response Relationship, Drug , Enzyme Inhibitors/toxicity , Glutathione/metabolism , Malonates/toxicity , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurons/metabolism , Neurons/pathology , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/antagonists & inhibitors , Succinate Dehydrogenase/metabolism
12.
Exp Neurol ; 196(1): 96-103, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16112113

ABSTRACT

The pathogenesis of non-glutamatergic, depolarization-induced cell death is still enigmatic. Recently, we have shown that veratridine induces apoptosis in chromaffin cells, and we have demonstrated protective effects of antioxidants in this system, suggesting a role for Na+ channels and oxidative stress in depolarization-induced cell death. We examined the possible contribution of p53, a transcription factor that has a major role in determining cell fate, and the mitochondrial apoptosis pathway in veratridine-induced cell death of cultured bovine chromaffin cells. Nuclear condensation and fragmentation were detected several hours after a 60-min exposure to 30 microM veratridine. Apoptosis was associated with a transitory increase in p53 protein levels. Veratridine induced transcription of the pro-apoptotic p53 target gene PUMA, but not of bax or pig3. Using transient transfection experiments, we found that wild-type p53, but not the mutant form p53-273H, was sufficient to induce cell death in the chromaffin cells, which was caspase-9 dependent. The down-regulation of either p53, by overexpressing p53-273H, or caspase-9 activity using a dominant-negative caspase-9 mutant protected chromaffin cells against veratridine-induced toxicity. Our data demonstrate the importance of p53 and the downstream activation of the mitochondrial apoptosis pathway in depolarization-induced apoptosis.


Subject(s)
Apoptosis/physiology , Chromaffin Cells/pathology , Genes, p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis/drug effects , Cattle , Cells, Cultured , Chromaffin Cells/drug effects , Electrophoresis, Polyacrylamide Gel , Genes, p53/drug effects , Immunoblotting , Immunohistochemistry , Transfection , Tumor Suppressor Proteins/drug effects , Veratridine/toxicity
13.
Neuroscience ; 133(4): 959-67, 2005.
Article in English | MEDLINE | ID: mdl-15964487

ABSTRACT

Minocycline, a semisynthetic derivative of tetracycline, displays beneficial activity in neuroprotective in models including, Parkinson disease, spinal cord injury, amyotrophic lateral sclerosis, Huntington disease and stroke. The mechanisms by which minocycline inhibits apoptosis remain poorly understood. In the present report we have investigated the effects of minocycline on mitochondria, due to their crucial role in apoptotic pathways. In mitochondria isolated suspensions, minocycline failed to block superoxide-induced swelling but was effective in blocking mitochondrial swelling induced by calcium. This latter effect might be mediated through dissipation of mitochondrial transmembrane potential and blockade of mitochondrial calcium uptake. Consistently, minocycline fails to protect SH-SY5Y cell cultures against reactive oxygen species-mediated cell death, including malonate and 6-hydroxydopamine treatments, but it is effective against staurosporine-induced cytotoxicity. The effects of this antibiotic on mitochondrial respiratory chain complex were also analyzed. Minocycline did not modify complex IV activity, and only at the higher concentration tested (100 microM) inhibited complex II/III activity. Other members of the minocycline antibiotic family like tetracycline failed to induce these mitochondrial effects.


Subject(s)
Calcium/metabolism , Membrane Potentials/drug effects , Minocycline/pharmacology , Mitochondria/drug effects , Mitochondrial Swelling/drug effects , Neuroprotective Agents/pharmacology , Animals , Calcium/pharmacology , Cell Count/methods , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Humans , NADP/metabolism , Neuroblastoma , Rats , Rats, Sprague-Dawley , Spectrophotometry/methods , Staurosporine/pharmacology , Tetrazolium Salts , Thiazoles
14.
Rev Neurol ; 39(3): 243-50, 2004.
Article in Spanish | MEDLINE | ID: mdl-15284966

ABSTRACT

INTRODUCTION: In this review we will study the role of protein p53 in neurodegenerative processes and conduct a detailed analysis of the mechanisms responsible for regulating its levels and biological activity. We analyse the neuropathologies in which this protein is involved, such as Alzheimer's and Parkinson's diseases and amyotrophic lateral sclerosis, and we will also examine its regulation by second messengers such as the reactive species of oxygen and calcium, showing the signalling paths involved in the apoptotic processes. DEVELOPMENT: The year 2004 sees the 25th anniversary of the discovery of protein p53. At first p53 was wrongly attributed with an oncogenic function due to its capacity to bind to the T antigen of the virus SV40 in transformed cells. Nevertheless, it was not until 1989 that it was attributed with its true physiological function as a tumour-suppressing protein. This milestone constitutes a turning point in the short life of this protein. Protein p53 plays a fundamental role in the mechanisms the cell uses to respond to damage or mutation in the genome. There is, therefore, a correlation between deletions or mutations in the p53 gene and the development of some kinds of cancer; additionally, increases in the protein levels of its native form have been reported in pathologies where apoptotic processes are high. CONCLUSIONS: Protein p53 plays an essential role in the mechanisms by which the cell responds to damage or mutation in the genome. It can activate two signalling mechanisms that lead either to stopping the cell cycle or to the death of the cell due to apoptosis if the cell cannot repair the damage to the genome. There is a correlation between its deletions and mutations and the development of cancer, and increases in its native form have been described in pathologies where apoptotic processes are high, as is the case of some neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/etiology , Tumor Suppressor Protein p53/physiology , Humans , Transcription Factors/physiology
15.
J Physiol Biochem ; 60(4): 287-307, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15957248

ABSTRACT

This year the p53 protein, also known as "guardian of the genome", turns twenty five years old. During this period the p53 knowledge have changed from an initial pro-oncogene activity to the tumorsupressor p53 function. p53 is activated upon stress signals, such as gamma irradiation, UV, hypoxia, virus infection, and DNA damage, leading to protection of cells by inducing target genes. The molecules activated by p53 induce cell cycle arrest, DNA repair to conserve the genome and apoptosis. The regulation of p53 functions is tightly controlled through several mechanisms including p53 transcription and translation, protein stability, post-translational modifications, and subcellular localization. In fact, mutations in p53 are the most frequent molecular alterations detected in human tumours. Furthermore, in some degenerative processes, fragmentation and oxidative damage in DNA take place, and in these situations p53 is involved. So, p53 is considered a pharmacological target, p53 overexpression induces apoptosis in cancer and its expression blockage protects cells against lethal insults.


Subject(s)
Genes, Tumor Suppressor/physiology , Genome, Human , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Humans , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...