Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Int J Mol Sci ; 24(2)2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36674792

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Animals , Humans , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/chemistry , Calcium/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Cell Death , Calcium Channels/genetics , Peptide Fragments/genetics , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Mammals/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism
2.
J Neurosci ; 36(46): 11693-11703, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27852777

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Models, Chemical , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Amyloid beta-Peptides/ultrastructure , Animals , Binding Sites , Cell Survival/physiology , Cells, Cultured , Computer Simulation , Mice , Models, Molecular , Neurons/cytology , Nitro Compounds/chemistry , Nitro Compounds/metabolism , Protein Binding , Protein Multimerization , Receptors, N-Methyl-D-Aspartate/chemistry , Tyrosine/chemistry , Tyrosine/metabolism
3.
Biochim Biophys Acta ; 1852(3): 421-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25500153

ABSTRACT

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.


Subject(s)
Apoptosis , Brain Ischemia/metabolism , Brain/metabolism , Fibrinogen/metabolism , Fibrinolysis , Neurons/metabolism , Stroke/metabolism , Adult , Aged , Aged, 80 and over , Animals , Brain/pathology , Brain Ischemia/pathology , Caspase 3/metabolism , Cell Line, Tumor , Enzyme Activation , Female , Humans , Male , Middle Aged , Neurons/pathology , Rats , Rats, Sprague-Dawley , Stroke/pathology , Tyrosine/analogs & derivatives , Tyrosine/metabolism
4.
Mol Membr Biol ; 31(5): 152-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25046533

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/ultrastructure , Brain/drug effects , Drug Delivery Systems , Endothelium, Vascular/ultrastructure , Blood-Brain Barrier/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Endothelium, Vascular/drug effects , Humans , Liposomes/administration & dosage , Mental Disorders/drug therapy , Mental Disorders/pathology , Nanotechnology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology
5.
PLoS One ; 13(3): e0194476, 2018.
Article in English | MEDLINE | ID: mdl-29554110

ABSTRACT

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.


Subject(s)
Cardiomegaly , Catechin/analogs & derivatives , Memory Disorders , Memory, Short-Term/drug effects , Williams Syndrome , Animals , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Catechin/pharmacology , Disease Models, Animal , Memory Disorders/drug therapy , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Mutant Strains , Williams Syndrome/drug therapy , Williams Syndrome/genetics , Williams Syndrome/metabolism , Williams Syndrome/pathology
6.
Oncotarget ; 7(12): 13354-71, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26968811

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis , Distal Myopathies/pathology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Myoblasts/pathology , N-Acetylneuraminic Acid/metabolism , Adult , Case-Control Studies , Cells, Cultured , Distal Myopathies/metabolism , Female , Humans , Male , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism
7.
Oncotarget ; 7(37): 58876-58892, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27557499

ABSTRACT

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.


Subject(s)
Cerebellar Cortex/pathology , Disks Large Homolog 4 Protein/metabolism , Eukaryotic Initiation Factor-2/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Eukaryotic Initiation Factor-2/genetics , Excitatory Amino Acid Agents/metabolism , Glutamic Acid/metabolism , Heme/metabolism , Humans , Memory , Mice , Mice, Inbred Strains , Neurons/pathology , Nitric Oxide/metabolism , Phosphorylation , Protein Biosynthesis , RNA, Small Interfering/genetics , Receptors, N-Methyl-D-Aspartate/genetics
8.
Antioxid Redox Signal ; 22(15): 1295-307, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25706765

ABSTRACT

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.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Synapses/metabolism , eIF-2 Kinase/metabolism , Animals , Cells, Cultured , Eukaryotic Initiation Factor-2/metabolism , Glutamic Acid/pharmacology , Hippocampus/embryology , Hippocampus/metabolism , Humans , Memory Consolidation , Mice , Neurons/cytology , Phosphorylation , Protein Biosynthesis , Rats
9.
Brain Res ; 1573: 1-16, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24854120

ABSTRACT

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.


Subject(s)
Actin Cytoskeleton/physiology , Brain/ultrastructure , Dendritic Spines/physiology , Glutamic Acid/metabolism , Actin Cytoskeleton/ultrastructure , Aging , Animals , Brain/physiology , Brain/physiopathology , Dendritic Spines/ultrastructure , Humans , Neuronal Plasticity
10.
Nutrients ; 6(8): 3000-17, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25079853

ABSTRACT

Niemann-Pick C (NPC) disease is a fatal neurodegenerative disorder characterized by the accumulation of free cholesterol in lysosomes. We have previously reported that oxidative stress is the main upstream stimulus activating the proapoptotic c-Abl/p73 pathway in NPC neurons. We have also observed accumulation of vitamin E in NPC lysosomes, which could lead to a potential decrease of its bioavailability. Our aim was to determine if dietary vitamin E supplementation could improve NPC disease in mice. NPC mice received an alpha-tocopherol (α-TOH) supplemented diet and neurological symptoms, survival, Purkinje cell loss, α-TOH and nitrotyrosine levels, astrogliosis, and the c-Abl/p73 pathway functions were evaluated. In addition, the effect of α-TOH on the c-Abl/p73 pathway was evaluated in an in vitro NPC neuron model. The α-TOH rich diet delayed loss of weight, improved coordination and locomotor function and increased the survival of NPC mice. We found increased Purkinje neurons and α-TOH levels and reduced astrogliosis, nitrotyrosine and phosphorylated p73 in cerebellum. A decrease of c-Abl/p73 activation was also observed in the in vitro NPC neurons treated with α-TOH. In conclusion, our results show that vitamin E can delay neurodegeneration in NPC mice and suggest that its supplementation in the diet could be useful for the treatment of NPC patients.


Subject(s)
DNA-Binding Proteins/metabolism , Dietary Supplements , Neurodegenerative Diseases/drug therapy , Niemann-Pick Disease, Type C/drug therapy , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Tumor Suppressor Proteins/metabolism , Vitamin E/administration & dosage , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , DNA-Binding Proteins/genetics , Disease Models, Animal , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Niemann-Pick Disease, Type C/genetics , Nuclear Proteins/genetics , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-abl/genetics , Signal Transduction , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism , alpha-Tocopherol
11.
Neurosci Lett ; 580: 78-82, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25102327

ABSTRACT

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.


Subject(s)
Caspase 3/metabolism , Membrane Potential, Mitochondrial , Neurons/metabolism , Pyruvaldehyde/metabolism , bcl-2-Associated X Protein/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Apoptosis , Cell Line, Tumor , Cell Survival , Glycosylation , Humans , Mutation , Neurons/drug effects , Neurons/pathology , Peptide Fragments/toxicity , Pyruvaldehyde/pharmacology , Triose-Phosphate Isomerase/genetics , Triose-Phosphate Isomerase/metabolism
12.
J Alzheimers Dis ; 41(1): 273-88, 2014.
Article in English | MEDLINE | ID: mdl-24614897

ABSTRACT

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.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Apoptosis/physiology , Neurons/physiology , Peptide Fragments/metabolism , Pyruvaldehyde/metabolism , Triose-Phosphate Isomerase/metabolism , Aged , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/genetics , Brain/physiopathology , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Computer Simulation , Female , Humans , Male , Mice, Transgenic , Middle Aged , Models, Molecular , Presenilin-1/genetics , Triose-Phosphate Isomerase/genetics
13.
J Alzheimers Dis ; 40(3): 643-57, 2014.
Article in English | MEDLINE | ID: mdl-24503620

ABSTRACT

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.


Subject(s)
Albumins/metabolism , Alzheimer Disease , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Protein Processing, Post-Translational/physiology , Tyrosine/analogs & derivatives , Aged , Albumins/drug effects , Albumins/pharmacology , Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Brain/cytology , Brain/metabolism , Brain/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Female , Glycosylation , Humans , Male , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Neurons/drug effects , Protein Aggregates/physiology , Trypsin/pharmacology , Tyrosine/metabolism , tau Proteins/metabolism
14.
Oxid Med Cell Longev ; 2013: 826143, 2013.
Article in English | MEDLINE | ID: mdl-23983901

ABSTRACT

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.


Subject(s)
Oxidative Stress/drug effects , Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrogen Peroxide/pharmacology , Mice , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Tyrosine/analogs & derivatives , Tyrosine/drug effects
15.
PLoS One ; 8(9): e74203, 2013.
Article in English | MEDLINE | ID: mdl-24069280

ABSTRACT

Calcium signaling in the brain is fundamental to the learning and memory process and there is evidence to suggest that its dysfunction is involved in the pathological pathways underlying Alzheimer's disease (AD). Recently, the calcium hypothesis of AD has received support with the identification of the non-selective Ca(2+)-permeable channel CALHM1. A genetic polymorphism (p. P86L) in CALHM1 reduces plasma membrane Ca(2+) permeability and is associated with an earlier age-at-onset of AD. To investigate the role of CALHM1 variants in early-onset AD (EOAD), we sequenced all CALHM1 coding regions in three independent series comprising 284 EOAD patients and 326 controls. Two missense mutations in patients (p.G330D and p.R154H) and one (p.A213T) in a control individual were identified. Calcium imaging analyses revealed that while the mutation found in a control (p.A213T) behaved as wild-type CALHM1 (CALHM1-WT), a complete abolishment of the Ca(2+) influx was associated with the mutations found in EOAD patients (p.G330D and p.R154H). Notably, the previously reported p. P86L mutation was associated with an intermediate Ca(2+) influx between the CALHM1-WT and the p.G330D and p.R154H mutations. Since neither expression of wild-type nor mutant CALHM1 affected amyloid ß-peptide (Aß) production or Aß-mediated cellular toxicity, we conclude that rare genetic variants in CALHM1 lead to Ca(2+) dysregulation and may contribute to the risk of EOAD through a mechanism independent from the classical Aß cascade.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Polymorphism, Genetic , Adult , Age of Onset , Aged , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Calcium Signaling , Case-Control Studies , DNA Mutational Analysis , Female , Homeostasis/genetics , Humans , Male , Middle Aged , Mutation , Sequence Alignment
16.
PLoS One ; 6(6): e21456, 2011.
Article in English | MEDLINE | ID: mdl-21738672

ABSTRACT

BACE1 is a key enzyme involved in the production of amyloid ß-peptide (Aß) in Alzheimer's disease (AD) brains. Normally, its expression is constitutively inhibited due to the presence of the 5'untranslated region (5'UTR) in the BACE1 promoter. BACE1 expression is activated by phosphorylation of the eukaryotic initiation factor (eIF)2-alpha, which reverses the inhibitory effect exerted by BACE1 5'UTR. There are four kinases associated with different types of stress that could phosphorylate eIF2-alpha. Here we focus on the double-stranded (ds) RNA-activated protein kinase (PKR). PKR is activated during viral infection, including that of herpes simplex virus type 1 (HSV1), a virus suggested to be implicated in the development of AD, acting when present in brains of carriers of the type 4 allele of the apolipoprotein E gene. HSV1 is a dsDNA virus but it has genes on both strands of the genome, and from these genes complementary RNA molecules are transcribed. These could activate BACE1 expression by the PKR pathway. Here we demonstrate in HSV1-infected neuroblastoma cells, and in peripheral nervous tissue from HSV1-infected mice, that HSV1 activates PKR. Cloning BACE1 5'UTR upstream of a luciferase (luc) gene confirmed its inhibitory effect, which can be prevented by salubrinal, an inhibitor of the eIF2-alpha phosphatase PP1c. Treatment with the dsRNA analog poly (I∶C) mimicked the stimulatory effect exerted by salubrinal over BACE1 translation in the 5'UTR-luc construct and increased Aß production in HEK-APPsw cells. Summarizing, our data suggest that PKR activated in brain by HSV1 could play an important role in the development of AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Herpesvirus 1, Human/physiology , eIF-2 Kinase/metabolism , Aged , Amyloid Precursor Protein Secretases/genetics , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Female , HeLa Cells , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred BALB C , eIF-2 Kinase/genetics
17.
J Alzheimers Dis ; 22(2): 641-52, 2010.
Article in English | MEDLINE | ID: mdl-20858976

ABSTRACT

Different mechanisms including oxidative stress are proposed for amyloid-ß peptide (Aß) neurotoxicity, and here we contribute to demonstrate that nitro-oxidative stress is playing a key role. Yeasts are a well-known model for H2O2 toxicity. Interestingly, yeast cell wall prevents interaction of Aß fibrils with membrane receptors or calcium channels and we found a significant viability reduction in yeasts when challenged with Aß fibrils. Furthermore, iron and copper chelators, as well as the antioxidants glutathione and trolox, were neuroprotective on neuroblastoma cells and mouse hippocampal neurons challenged with Aß fibrils. Glutathione prevents the oxidation, glycation and nitrotyrosination of cell proteins induced by Aß. Trolox protected neurons in cell viability studies, maintaining the vesicular transport integrity and preventing the trigger of apoptotic mechanisms. Interestingly, we have also found that brain derived neuronal factor (BDNF) and neurotrophin-3 (NT-3) were able to protect mouse hippocampal and cortical neurons against H2O2 and Aß fibrils. Considering that superoxide anion, produced by Aß cell damage, and nitric oxide, whose production is altered in AD, react to form the highly reactive peroxynitrite anion, we studied the role of trolox to ameliorate the peroxynitrite cell damage. Finally, one of the major proteins to be nitrotyrosinated in AD, the triose phosphate isomerase (TPI) was assayed searching for a denitrase activity that could reverse intracellular nitrotyrosination. We have found that human neuroblastoma SH-SY5Y cells express a constitutive denitrase activity that partially denitrated nitro-TPI. Altogether, our results support a key role of nitro-oxidative stress in the neuronal damage induced by Aß fibrils.


Subject(s)
Amyloid beta-Peptides/pharmacology , Amyloid/metabolism , Neurons/drug effects , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Aged , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Survival , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chromans/pharmacology , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian , Glutathione/metabolism , Hippocampus/cytology , Humans , Hydrogen Peroxide/pharmacology , Immunoprecipitation/methods , In Situ Nick-End Labeling/methods , Male , Mice , Models, Biological , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Peptide Fragments/pharmacology , Rats , Siderophores/pharmacology , Thymus Extracts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL