Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Mol Sci ; 24(2)2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36674792

RESUMEN

Alzheimer's disease (AD) is known to be caused by amyloid ß-peptide (Aß) misfolded into ß-sheets, but this knowledge has not yet led to treatments to prevent AD. To identify novel molecular players in Aß toxicity, we carried out a genome-wide screen in Saccharomyces cerevisiae, using a library of 5154 gene knock-out strains expressing Aß1-42. We identified 81 mammalian orthologue genes that enhance Aß1-42 toxicity, while 157 were protective. Next, we performed interactome and text-mining studies to increase the number of genes and to identify the main cellular functions affected by Aß oligomers (oAß). We found that the most affected cellular functions were calcium regulation, protein translation and mitochondrial activity. We focused on SURF4, a protein that regulates the store-operated calcium channel (SOCE). An in vitro analysis using human neuroblastoma cells showed that SURF4 silencing induced higher intracellular calcium levels, while its overexpression decreased calcium entry. Furthermore, SURF4 silencing produced a significant reduction in cell death when cells were challenged with oAß1-42, whereas SURF4 overexpression induced Aß1-42 cytotoxicity. In summary, we identified new enhancer and protective activities for Aß toxicity and showed that SURF4 contributes to oAß1-42 neurotoxicity by decreasing SOCE activity.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Animales , Humanos , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/química , Calcio/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Muerte Celular , Canales de Calcio/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/metabolismo , Mamíferos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
2.
J Neurosci ; 36(46): 11693-11703, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27852777

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the pathological aggregation of the amyloid-ß peptide (Aß). Monomeric soluble Aß can switch from helicoidal to ß-sheet conformation, promoting its assembly into oligomers and subsequently to amyloid fibrils. Oligomers are highly toxic to neurons and have been reported to induce synaptic transmission impairments. The progression from oligomers to fibrils forming senile plaques is currently considered a protective mechanism to avoid the presence of the highly toxic oligomers. Protein nitration is a frequent post-translational modification under AD nitrative stress conditions. Aß can be nitrated at tyrosine 10 (Y10) by peroxynitrite. Based on our analysis of ThT binding, Western blot and electron and atomic force microscopy, we report that Aß nitration stabilizes soluble, highly toxic oligomers and impairs the formation of fibrils. We propose a mechanism by which fibril elongation is interrupted upon Y10 nitration: Nitration disrupts fibril-forming folds by preventing H14-mediated bridging, as shown with an Aß analog containing a single residue (H to E) replacement that mimics the behavior of nitrated Aß related to fibril formation and neuronal toxicity. The pathophysiological role of our findings in AD was highlighted by the study of these nitrated oligomers on mouse hippocampal neurons, where an increased NMDAR-dependent toxicity of nitrated Aß oligomers was observed. Our results show that Aß nitrotyrosination is a post-translational modification that increases Aß synaptotoxicity. SIGNIFICANCE STATEMENT: We report that nitration (i.e., the irreversible addition of a nitro group) of the Alzheimer-related peptide amyloid-ß (Aß) favors the stabilization of highly toxic oligomers and inhibits the formation of Aß fibrils. The nitrated Aß oligomers are more toxic to neurons due to increased cytosolic calcium levels throughout their action on NMDA receptors. Sustained elevated calcium levels trigger excitotoxicity, a characteristic event in Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Modelos Químicos , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Amiloide/química , Amiloide/ultraestructura , Péptidos beta-Amiloides/ultraestructura , Animales , Sitios de Unión , Supervivencia Celular/fisiología , Células Cultivadas , Simulación por Computador , Ratones , Modelos Moleculares , Neuronas/citología , Nitrocompuestos/química , Nitrocompuestos/metabolismo , Unión Proteica , Multimerización de Proteína , Receptores de N-Metil-D-Aspartato/química , Tirosina/química , Tirosina/metabolismo
3.
Biochim Biophys Acta ; 1852(3): 421-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25500153

RESUMEN

Ischemic stroke is an acute vascular event that compromises neuronal viability, and identification of the pathophysiological mechanisms is critical for its correct management. Ischemia produces increased nitric oxide synthesis to recover blood flow but also induces a free radical burst. Nitric oxide and superoxide anion react to generate peroxynitrite that nitrates tyrosines. We found that fibrinogen nitrotyrosination was detected in plasma after the initiation of ischemic stroke in human patients. Electron microscopy and protein intrinsic fluorescence showed that in vitro nitrotyrosination of fibrinogen affected its structure. Thromboelastography showed that initially fibrinogen nitrotyrosination retarded clot formation but later made the clot more resistant to fibrinolysis. This result was independent of any effect on thrombin production. Immunofluorescence analysis of affected human brain areas also showed that both fibrinogen and nitrotyrosinated fibrinogen spread into the brain parenchyma after ischemic stroke. Therefore, we assayed the toxicity of fibrinogen and nitrotyrosinated fibrinogen in a human neuroblastoma cell line. For that purpose we measured the activity of caspase-3, a key enzyme in the apoptotic pathway, and cell survival. We found that nitrotyrosinated fibrinogen induced higher activation of caspase 3. Accordingly, cell survival assays showed a more neurotoxic effect of nitrotyrosinated fibrinogen at all concentrations tested. In summary, nitrotyrosinated fibrinogen would be of pathophysiological interest in ischemic stroke due to both its impact on hemostasis - it impairs thrombolysis, the main target in stroke treatments - and its neurotoxicity that would contribute to the death of the brain tissue surrounding the infarcted area.


Asunto(s)
Apoptosis , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Fibrinógeno/metabolismo , Fibrinólisis , Neuronas/metabolismo , Accidente Cerebrovascular/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/patología , Isquemia Encefálica/patología , Caspasa 3/metabolismo , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/patología , Tirosina/análogos & derivados , Tirosina/metabolismo
4.
Mol Membr Biol ; 31(5): 152-67, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25046533

RESUMEN

The blood-brain barrier (BBB) is constituted by a specialized vascular endothelium that interacts directly with astrocytes, neurons and pericytes. It protects the brain from the molecules of the systemic circulation but it has to be overcome for the proper treatment of brain cancer, psychiatric disorders or neurodegenerative diseases, which are dramatically increasing as the population ages. In the present work we have revised the current knowledge on the cellular structure of the BBB and the different procedures utilized currently and those proposed to cross it. Chemical modifications of the drugs, such as increasing their lipophilicity, turn them more prone to be internalized in the brain. Other mechanisms are the use of molecular tools to bind the drugs such as small immunoglobulins, liposomes or nanoparticles that will act as Trojan Horses favoring the drug delivery in brain. This fusion of the classical pharmacology with nanotechnology has opened a wide field to many different approaches with promising results to hypothesize that BBB will not be a major problem for the new generation of neuroactive drugs. The present review provides an overview of all state-of-the-art of the BBB structure and function, as well as of the classic strategies and these appeared in recent years to deliver drugs into the brain for the treatment of Central Nervous System (CNS) diseases.


Asunto(s)
Barrera Hematoencefálica/ultraestructura , Encéfalo/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Endotelio Vascular/ultraestructura , Barrera Hematoencefálica/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Endotelio Vascular/efectos de los fármacos , Humanos , Liposomas/administración & dosificación , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/patología , Nanotecnología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología
5.
PLoS One ; 13(3): e0194476, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29554110

RESUMEN

Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder caused by a heterozygous deletion of 26-28 genes at chromosome band 7q11.23. The complete deletion (CD) mouse model mimics the most common deletion found in WBS patients and recapitulates most neurologic features of the disorder along with some cardiovascular manifestations leading to significant cardiac hypertrophy with increased cardiomyocytes' size. Epigallocatechin-3-gallate (EGCG), the most abundant catechin found in green tea, has been associated with potential health benefits, both on cognition and cardiovascular phenotypes, through several mechanisms. We aimed to investigate the effects of green tea extracts on WBS-related phenotypes through a phase I clinical trial in mice. After feeding CD animals with green tea extracts dissolved in the drinking water, starting at three different time periods (prenatal, youth and adulthood), a set of behavioral tests and several anatomical, histological and molecular analyses were performed. Treatment resulted to be effective in the reduction of cardiac hypertrophy and was also able to ameliorate short-term memory deficits of CD mice. Taken together, these results suggest that EGCG might have a therapeutic and/or preventive role in the management of WBS.


Asunto(s)
Cardiomegalia , Catequina/análogos & derivados , Trastornos de la Memoria , Memoria a Corto Plazo/efectos de los fármacos , Síndrome de Williams , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Catequina/farmacología , Modelos Animales de Enfermedad , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones Mutantes , Síndrome de Williams/tratamiento farmacológico , Síndrome de Williams/genética , Síndrome de Williams/metabolismo , Síndrome de Williams/patología
6.
Oncotarget ; 8(25): 41154-41165, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28467807

RESUMEN

The amyloid beta-peptide (Aß) plays a leading role in Alzheimer's disease (AD) physiopathology. Even though monomeric forms of Aß are harmless to cells, Aß can aggregate into ß-sheet oligomers and fibrils, which are both neurotoxic. Therefore, one of the main therapeutic approaches to cure or delay AD onset and progression is targeting Aß aggregation. In the present study, we show that a pool of human gamma immunoglobulins (IgG) protected cortical neurons from the challenge with Aß oligomers, as assayed by MTT reduction, caspase-3 activation and cytoskeleton integrity. In addition, we report the inhibitory effect of IgG on Aß aggregation, as shown by Thioflavin T assay, size exclusion chromatography and atomic force microscopy. Similar results were obtained with Palivizumab, a human anti-sincitial virus antibody. In order to dissect the important domains, we cleaved the pool of human IgG with papain to obtain Fab and Fc fragments. Using these cleaved fragments, we functionally identified Fab as the immunoglobulin fragment inhibiting Aß aggregation, a result that was further confirmed by an in silico structural model. Interestingly, bioinformatic tools show a highly conserved structure able to bind amyloid in the Fab region. Overall, our data strongly support the inhibitory effect of human IgG on Aß aggregation and its neuroprotective role.


Asunto(s)
Péptidos beta-Amiloides/química , Cadenas gamma de Inmunoglobulina/farmacología , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Antígenos/metabolismo , Humanos , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/metabolismo , Fragmentos de Inmunoglobulinas/farmacología , Cadenas gamma de Inmunoglobulina/química , Cadenas gamma de Inmunoglobulina/metabolismo , Modelos Moleculares , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/prevención & control , Unión Proteica
7.
Oncotarget ; 7(12): 13354-71, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26968811

RESUMEN

GNE myopathy is an autosomal recessive muscular disorder of young adults characterized by progressive skeletal muscle weakness and wasting. It is caused by a mutation in the UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) gene, which encodes a key enzyme in sialic acid biosynthesis. The mutated hypofunctional GNE is associated with intracellular accumulation of amyloid ß-peptide (Aß) in patient muscles through as yet unknown mechanisms. We found here for the first time that an experimental reduction in sialic acid favors Aß1-42 endocytosis in C2C12 myotubes, which is dependent on clathrin and heparan sulfate proteoglycan. Accordingly, Aß1-42 internalization in myoblasts from a GNE myopathy patient was enhanced. Next, we investigated signal changes triggered by Aß1-42 that may underlie toxicity. We observed that p-Akt levels are reduced in step with an increase in apoptotic markers in GNE myopathy myoblasts compared to control myoblasts. The same results were experimentally obtained when Aß1-42 was overexpressed in myotubes. Hence, we propose a novel disease mechanism whereby hyposialylation favors Aß1-42 internalization and the subsequent apoptosis in myotubes and in skeletal muscle from GNE myopathy patients.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apoptosis , Miopatías Distales/patología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Mioblastos/patología , Ácido N-Acetilneuramínico/metabolismo , Adulto , Estudios de Casos y Controles , Células Cultivadas , Miopatías Distales/metabolismo , Femenino , Humanos , Masculino , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo
8.
Oncotarget ; 7(37): 58876-58892, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27557499

RESUMEN

The activation of N-Methyl D-Aspartate Receptor (NMDAR) by glutamate is crucial in the nervous system function, particularly in memory and learning. NMDAR is composed by two GluN1 and two GluN2 subunits. GluN2B has been reported to participate in the prevalent NMDAR subtype at synapses, the GluN1/2A/2B. Here we studied the regulation of GluN2B expression in cortical neurons finding that glutamate up-regulates GluN2B translation through the action of nitric oxide (NO), which induces the phosphorylation of the eukaryotic translation initiation factor 2 α (eIF2α). It is a process mediated by the NO-heme-regulated eIF2α kinase (HRI), as the effect was avoided when a specific HRI inhibitor or a HRI small interfering RNA (siHRI) were used. We found that the expressed GluN2B co-localizes with PSD-95 at the postsynaptic ending, which strengthen the physiological relevance of the proposed mechanism. Moreover the receptors bearing GluN2B subunits upon NO stimulation are functional as high Ca2+ entry was measured and increases the co-localization between GluN2B and GluN1 subunits. In addition, the injection of the specific HRI inhibitor in mice produces a decrease in memory retrieval as tested by the Novel Object Recognition performance. Summarizing our data suggests that glutamatergic stimulation induces HRI activation by NO to trigger GluN2B expression and this process would be relevant to maintain postsynaptic activity in cortical neurons.


Asunto(s)
Corteza Cerebelosa/patología , Homólogo 4 de la Proteína Discs Large/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células Cultivadas , Factor 2 Eucariótico de Iniciación/genética , Fármacos actuantes sobre Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Hemo/metabolismo , Humanos , Memoria , Ratones , Ratones Endogámicos , Neuronas/patología , Óxido Nítrico/metabolismo , Fosforilación , Biosíntesis de Proteínas , ARN Interferente Pequeño/genética , Receptores de N-Metil-D-Aspartato/genética
9.
Antioxid Redox Signal ; 22(15): 1295-307, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25706765

RESUMEN

AIMS: Hippocampus is the brain center for memory formation, a process that requires synaptogenesis. However, hippocampus is dramatically compromised in Alzheimer's disease due to the accumulation of amyloid ß-peptide, whose production is initiated by ß-site APP Cleaving Enzyme 1 (BACE1). It is known that pathological stressors activate BACE1 translation through the phosphorylation of the eukaryotic initiation factor-2α (eIF2α) by GCN2, PERK, or PKR kinases, leading to amyloidogenesis. However, BACE1 physiological regulation is still unclear. Since nitric oxide (NO) participates directly in hippocampal glutamatergic signaling, we investigated the neuronal role of the heme-regulated eukaryotic initiation factor eIF2α kinase (HRI), which can bind NO by a heme group, in BACE1 translation and its physiological consequences. RESULTS: We found that BACE1 is expressed on glutamate activation with NO being the downstream effector by triggering eIF2α phosphorylation, as it was obtained by Western blot and luciferase assay. It is due to the activation of HRI by NO as assayed by Western blot and immunofluorescence with an HRI inhibitor and HRI siRNA. BACE1 expression was early detected at synaptic spines, contributing to spine growth and consolidating the hippocampal memory as assayed with mice treated with HRI or neuronal NO synthase inhibitors. INNOVATION: We provide the first description that HRI and eIF2α are working in physiological conditions in the brain under the control of nitric oxide and glutamate signaling, and also that BACE1 has a physiological role in hippocampal function. CONCLUSION: We conclude that BACE1 translation is controlled by NO through HRI in glutamatergic hippocampal synapses, where it plays physiological functions, allowing the spine growth and memory consolidation.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Sinapsis/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Células Cultivadas , Factor 2 Eucariótico de Iniciación/metabolismo , Ácido Glutámico/farmacología , Hipocampo/embriología , Hipocampo/metabolismo , Humanos , Consolidación de la Memoria , Ratones , Neuronas/citología , Fosforilación , Biosíntesis de Proteínas , Ratas
10.
Brain Res ; 1573: 1-16, 2014 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-24854120

RESUMEN

Dendritic spines are actin-rich protrusions from the dendritic shaft, considered to be the locus where most synapses occur, as they receive the vast majority of excitatory connections in the central nervous system (CNS). Interestingly, hippocampal spines are plastic structures that contain a dense array of molecules involved in postsynaptic signaling and synaptic plasticity. Since changes in spine shape and size are correlated with the strength of excitatory synapses, spine morphology directly reflects spine function. Therefore several neuropathologies are associated with defects in proteins located at the spines. The present work is focused on the spine actin cytoskeleton attending to its structure and function mainly in glutamatergic neurons. It addresses the study of the structural plasticity of dendritic spines associated with long-term potentiation (LTP) and the mechanisms that underlie learning and memory formation. We have integrated the current knowledge on synaptic proteins to relate this plethora of molecules with actin and actin-binding proteins. We further included recent findings that outline key uncharacterized proteins that would be useful to unveil the real ultrastructure and function of dendritic spines. Furthermore, this review is directed to understand how such spine diversity and interplay contributes to the regulation of spine morphogenesis and dynamics. It highlights their physiological relevance in the brain function, as well as it provides insights for pathological processes affecting dramatically dendritic spines, such as Alzheimer's disease.


Asunto(s)
Citoesqueleto de Actina/fisiología , Encéfalo/ultraestructura , Espinas Dendríticas/fisiología , Ácido Glutámico/metabolismo , Citoesqueleto de Actina/ultraestructura , Envejecimiento , Animales , Encéfalo/fisiología , Encéfalo/fisiopatología , Espinas Dendríticas/ultraestructura , Humanos , Plasticidad Neuronal
11.
Neurosci Lett ; 580: 78-82, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25102327

RESUMEN

Alzheimer's disease (AD) is characterized by the oxidative stress generated from amyloid ß-peptide (Aß) aggregates. It produces protein nitrotyrosination, after the reaction with nitric oxide to form peroxynitrite, being triosephosphate isomerase (TPI) one of the most affected proteins. TPI is a glycolytic enzyme that catalyzes the interconversion between glyceraldehyde 3-phosphate (GAP) and dihydroxyacetone phosphate (DHAP). Methylglyoxal (MG) is a by-product of TPI activity whose production is triggered when TPI is nitrotyrosinated. MG is harmful to cells because it glycates proteins. Here we found protein glycation when human neuroblastoma cells were treated with Aß. Moreover glycation was also observed when neuroblastoma cells overexpressed mutated TPI where Tyr165 or Tyr209, the two tyrosines close to the catalytic center, were changed by Phe in order to mimic the effect of nitrotyrosination. The pathological relevance of these findings was studied by challenging cells with Aß oligomers and MG. A significant decrease in mitochondrial transmembrane potential, one of the first apoptotic events, was obtained. Therefore, increasing concentrations of MG were assayed searching for MG effect in neuronal apoptosis. We found a decrease of the protective Bcl2 and an increase of the proapoptotic caspase-3 and Bax levels. Our results suggest that MG is triggering apoptosis in neurons and it would play a key role in AD neurodegeneration.


Asunto(s)
Caspasa 3/metabolismo , Potencial de la Membrana Mitocondrial , Neuronas/metabolismo , Piruvaldehído/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/toxicidad , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Glicosilación , Humanos , Mutación , Neuronas/efectos de los fármacos , Neuronas/patología , Fragmentos de Péptidos/toxicidad , Piruvaldehído/farmacología , Triosa-Fosfato Isomerasa/genética , Triosa-Fosfato Isomerasa/metabolismo
12.
J Alzheimers Dis ; 41(1): 273-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24614897

RESUMEN

Amyloid-ß peptide (Aß) aggregates induce nitro-oxidative stress, contributing to the characteristic neurodegeneration found in Alzheimer's disease (AD). One of the most strongly nitrotyrosinated proteins in AD is the triosephosphate isomerase (TPI) enzyme which regulates glycolytic flow, and its efficiency decreased when it is nitrotyrosinated. The main aims of this study were to analyze the impact of TPI nitrotyrosination on cell viability and to identify the mechanism behind this effect. In human neuroblastoma cells (SH-SY5Y), we evaluated the effects of Aß42 oligomers on TPI nitrotyrosination. We found an increased production of methylglyoxal (MG), a toxic byproduct of the inefficient nitro-TPI function. The proapoptotic effects of Aß42 oligomers, such as decreasing the protective Bcl2 and increasing the proapoptotic caspase-3 and Bax, were prevented with a MG chelator. Moreover, we used a double mutant TPI (Y165F and Y209F) to mimic nitrosative modifications due to Aß action. Neuroblastoma cells transfected with the double mutant TPI consistently triggered MG production and a decrease in cell viability due to apoptotic mechanisms. Our data show for the first time that MG is playing a key role in the neuronal death induced by Aß oligomers. This occurs because of TPI nitrotyrosination, which affects both tyrosines associated with the catalytic center.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Apoptosis/fisiología , Neuronas/fisiología , Fragmentos de Péptidos/metabolismo , Piruvaldehído/metabolismo , Triosa-Fosfato Isomerasa/metabolismo , Anciano , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/genética , Encéfalo/fisiopatología , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Simulación por Computador , Femenino , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Modelos Moleculares , Presenilina-1/genética , Triosa-Fosfato Isomerasa/genética
13.
J Alzheimers Dis ; 40(3): 643-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24503620

RESUMEN

Glycation and nitrotyrosination are pathological posttranslational modifications that make proteins prone to losing their physiological properties. Since both modifications are increased in Alzheimer's disease (AD) due to amyloid-ß peptide (Aß) accumulation, we have studied their effect on albumin, the most abundant protein in cerebrospinal fluid and blood. Brain and plasmatic levels of glycated and nitrated albumin were significantly higher in AD patients than in controls. In vitro turbidometry and electron microscopy analyses demonstrated that glycation and nitrotyrosination promote changes in albumin structure and biochemical properties. Glycated albumin was more resistant to proteolysis and less uptake by hepatoma cells occurred. Glycated albumin also reduced the osmolarity expected for a solution containing native albumin. Both glycation and nitrotyrosination turned albumin cytotoxic in a cell type-dependent manner for cerebral and vascular cells. Finally, of particular relevance to AD, these modified albumins were significantly less effective in avoiding Aß aggregation than native albumin. In summary, nitrotyrosination and especially glycation alter albumin structural and biochemical properties, and these modifications might contribute for the progression of AD.


Asunto(s)
Albúminas/metabolismo , Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Tirosina/análogos & derivados , Anciano , Albúminas/efectos de los fármacos , Albúminas/farmacología , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/patología , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Femenino , Glicosilación , Humanos , Masculino , Molsidomina/análogos & derivados , Molsidomina/farmacología , Neuronas/efectos de los fármacos , Agregado de Proteínas/fisiología , Tripsina/farmacología , Tirosina/metabolismo , Proteínas tau/metabolismo
14.
Oxid Med Cell Longev ; 2013: 826143, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23983901

RESUMEN

Ischemic stroke is an acute vascular event that obstructs blood supply to the brain, producing irreversible damage that affects neurons but also glial and brain vessel cells. Immediately after the stroke, the ischemic tissue produces nitric oxide (NO) to recover blood perfusion but also produces superoxide anion. These compounds interact, producing peroxynitrite, which irreversibly nitrates protein tyrosines. The present study measured NO production in a human neuroblastoma (SH-SY5Y), a murine glial (BV2), a human endothelial cell line (HUVEC), and in primary cultures of human cerebral myocytes (HC-VSMCs) after experimental ischemia in vitro. Neuronal, endothelial, and inducible NO synthase (NOS) expression was also studied up to 24 h after ischemia, showing a different time course depending on the NOS type and the cells studied. Finally, we carried out cell viability experiments on SH-SY5Y cells with H2O2, a prooxidant agent, and with a NO donor to mimic ischemic conditions. We found that both compounds were highly toxic when they interacted, producing peroxynitrite. We obtained similar results when all cells were challenged with peroxynitrite. Our data suggest that peroxynitrite induces cell death and is a very harmful agent in brain ischemia.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Proteínas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Tirosina/análogos & derivados , Tirosina/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA