Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 23(15): 4688-4698, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26078011

ABSTRACT

Some symmetrical and unsymmetrical thiacarbocyanines bearing NO-donor nitrooxy and furoxan moieties were synthesized and studied as candidate anti-Alzheimer's drugs. All products activated soluble guanylate cyclase (sGC) in a dose-dependent manner, depending on the presence in their structures of NO-donor groups. None displayed toxicity when tested at concentrations below 10 µM on human brain microvascular endothelial cells (hCMEC/D3). Some products were capable of inhibiting amyloid ß-protein (Aß) aggregation, with a potency in the low µM concentration range, and of inhibiting aggregation of human recombinant tau protein in amyloid fibrils when incubated with the protein at 1 µM concentration. Nitrooxy derivative 21 and furoxan derivative 22 were selected to investigate synaptic plasticity. Both products, tested at 2 µM concentration, counteracted the inhibition of long-term potentiation (LTP) induced by Aß42 in hippocampal brain slices.


Subject(s)
Alzheimer Disease/drug therapy , Carbocyanines/therapeutic use , Nitric Oxide Donors/therapeutic use , Humans
2.
PLoS One ; 10(1): e0115352, 2015.
Article in English | MEDLINE | ID: mdl-25590583

ABSTRACT

A key element to delineate the biology of individual tumors is the regulation of apoptosis. In this work, we functionally characterize two breast cancer associated genes, the proteasome 26S subunit ATPase 3 interacting protein (PSMC3IP) and the epithelial-stromal interaction 1 (EPSTI1), to explore their potential apoptotic role in breast cancer. We first explore the existence of direct physical interactions with annotated BC-apoptotic genes. Based on the generated interaction network, we examine several apoptotic markers to determine the effect of PSMC3IP and EPSTI1 gene expression modulation in two different human breast cancer cell lines to suggest potential molecular mechanisms to unveil their role in the disease. Our results show that PSMC3IP and EPSTI1 are able to modulate the extrinsic apoptotic pathway in estrogen receptor positive and triple negative breast cancer cell lines, highlighting them as potential therapeutic targets.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans
3.
J Neurochem ; 128(2): 330-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117942

ABSTRACT

Autosomal-dominant Alzheimer's disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studying the mechanisms underlying these mutations can provide insight into the pathways that lead to AD pathology. The majority of biochemical studies on APP mutations to-date have focused on comparing mechanisms between mutations at different codons. It has been assumed that amino acid position is a major determinant of protein dysfunction and clinical phenotype. However, the differential effect of mutations at the same codon has not been sufficiently addressed. In the present study we compared the effects of the aggressive ADAD-associated APP I716F mutation with I716V and I716T on APP processing in human neuroglioma and CHO-K1 cells. All APP I716 mutations increased the ratio of Aß42/40 and changed the product line preference of γ-secretase towards Aß38 production. In addition, the APP I716F mutation impaired the ε-cleavage and the fourth cleavage of γ-secretase and led to abnormal APP ß-CTF accumulation at the plasma membrane. Taken together, these data indicate that APP mutations at the same codon can induce diverse abnormalities in APP processing, some resembling PSEN1 mutations. These differential effects could explain the clinical differences observed among ADAD patients bearing different APP mutations at the same position. The amyloid precursor protein (APP) I716F mutation is associated with autosomal dominant Alzheimer's disease with the youngest age-at-onset for the APP locus. Here, we describe that this mutation, when compared to two other familial Alzheimer's disease mutations at the same codon (I716V and I716T), interfered distinctly with γ-secretase cleavage. While all three mutations direct γ-secretase cleavage towards the 48→38 production line, the APP I716F mutation also impaired the ε-cleavage and the fourth cleavage of γ-secretase, resembling a PSEN1 mutation. These features may contribute to the aggressiveness of this mutation.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Adult , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Codon , Cricetinae , Cricetulus , Humans , Mutation
4.
PLoS One ; 8(3): e58837, 2013.
Article in English | MEDLINE | ID: mdl-23520537

ABSTRACT

A key event in the pathogenesis of Alzheimer's disease (AD) is the accumulation of amyloid-ß (Aß) species in the brain, derived from the sequential cleavage of the amyloid precursor protein (APP) by ß- and γ-secretases. Based on a systems biology study to repurpose drugs for AD, we explore the effect of lansoprazole, and other proton-pump inhibitors (PPIs), on Aß production in AD cellular and animal models. We found that lansoprazole enhances Aß37, Aß40 and Aß42 production and lowers Aß38 levels on amyloid cell models. Interestingly, acute lansoprazole treatment in wild type and AD transgenic mice promoted higher Aß40 levels in brain, indicating that lansoprazole may also exacerbate Aß production in vivo. Overall, our data presents for the first time that PPIs can affect amyloid metabolism, both in vitro and in vivo.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/biosynthesis , Enzyme Inhibitors/pharmacology , Proton Pump Inhibitors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Female , Humans , Lansoprazole , Mice , Mice, Knockout
5.
Acta Neuropathol ; 125(2): 201-13, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23224319

ABSTRACT

Autosomal-dominant Alzheimer disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studies from families with ADAD have been critical to support the amyloid cascade hypothesis of Alzheimer disease (AD), the basis for the current development of amyloid-based disease-modifying therapies in sporadic AD (SAD). However, whether the pathological changes in APP processing in the CNS in ADAD are similar to those observed in SAD remains unclear. In this study, we measured ß-site APP-cleaving enzyme (BACE) protein levels and activity, APP and APP C-terminal fragments in brain samples from subjects with ADAD carrying APP or PSEN1 mutations (n = 18), patients with SAD (n = 27) and age-matched controls (n = 22). We also measured sAPPß and BACE protein levels, as well as BACE activity, in CSF from individuals carrying PSEN1 mutations (10 mutation carriers and 7 non-carrier controls), patients with SAD (n = 32) and age-matched controls (n = 11). We found that in the brain, the pattern in ADAD was characterized by an increase in APP ß-C-terminal fragment (ß-CTF) levels despite no changes in BACE protein levels or activity. In contrast, the pattern in SAD in the brain was mainly characterized by an increase in BACE levels and activity, with less APP ß-CTF accumulation than ADAD. In the CSF, no differences were found between groups in BACE activity or expression or sAPPß levels. Taken together, these data suggest that the physiopathological events underlying the chronic Aß production/clearance imbalance in SAD and ADAD are different. These differences should be considered in the design of intervention trials in AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Central Nervous System/metabolism , Adult , Aged , Alzheimer Disease/cerebrospinal fluid , Amyloid Precursor Protein Secretases/cerebrospinal fluid , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/cerebrospinal fluid , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Aspartic Acid Endopeptidases/cerebrospinal fluid , Aspartic Acid Endopeptidases/metabolism , Blotting, Western , Female , Heterozygote , Humans , Immunohistochemistry , Male , Middle Aged , Mutation/genetics , Neurites/pathology , Presenilin-1/cerebrospinal fluid , Presenilin-1/genetics
6.
Bioessays ; 34(7): 532-41, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22513506

ABSTRACT

Here we postulate that the adapter protein evolutionarily conserved signalling intermediate in Toll pathway (ECSIT) might act as a molecular sensor in the pathogenesis of Alzheimer's disease (AD). Based on the analysis of our AD-associated protein interaction network, ECSIT emerges as an integrating signalling hub that ascertains cell homeostasis by the specific activation of protective molecular mechanisms in response to signals of amyloid-beta or oxidative damage. This converges into a complex cascade of patho-physiological processes. A failure to repair would generate severe mitochondrial damage and ultimately activate pro-apoptotic mechanisms, promoting synaptic dysfunction and neuronal death. Further support for our hypothesis is provided by increasing evidence of mitochondrial dysfunction in the disease etiology. Our model integrates seemingly controversial hypotheses for familial and sporadic forms of AD and envisions ECSIT as a biomarker to guide future therapies to halt or prevent AD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/pathology , Mitochondria/pathology , Oxidative Stress , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Age of Onset , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Apoptosis , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Genetic Predisposition to Disease , Homeostasis , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Mitochondria/immunology , Mitochondria/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/genetics , Presenilin-2/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Signal Transduction
7.
J Biol Chem ; 287(14): 11351-62, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22294685

ABSTRACT

NMDA receptor (NMDAR) stimulation promotes neuronal survival during brain development. Cerebellar granule cells (CGCs) need NMDAR stimulation to survive and develop. These neurons differentiate and mature during its migration from the external granular layer to the internal granular layer, and lack of excitatory inputs triggers their apoptotic death. It is possible to mimic this process in vitro by culturing CGCs in low KCl concentrations (5 mm) in the presence or absence of NMDA. Using this experimental approach, we have obtained whole genome expression profiles after 3 and 8 h of NMDA addition to identify genes involved in NMDA-mediated survival of CGCs. One of the identified genes was Nurr1, a member of the orphan nuclear receptor subfamily Nr4a. Our results report a direct regulation of Nurr1 by CREB after NMDAR stimulation. ChIP assay confirmed CREB binding to Nurr1 promoter, whereas CREB shRNA blocked NMDA-mediated increase in Nurr1 expression. Moreover, we show that Nurr1 is important for NMDAR survival effect. We show that Nurr1 binds to Bdnf promoter IV and that silencing Nurr1 by shRNA leads to a decrease in brain-derived neurotrophic factor (BDNF) protein levels and a reduction of NMDA neuroprotective effect. Also, we report that Nurr1 and BDNF show a similar expression pattern during postnatal cerebellar development. Thus, we conclude that Nurr1 is a downstream target of CREB and that it is responsible for the NMDA-mediated increase in BDNF, which is necessary for the NMDA-mediated prosurvival effect on neurons.


Subject(s)
Neurons/cytology , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Silencing , N-Methylaspartate/pharmacology , Neurons/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Rats , Rats, Wistar , Transcriptional Activation/drug effects
8.
PLoS One ; 6(10): e26609, 2011.
Article in English | MEDLINE | ID: mdl-22039514

ABSTRACT

BACKGROUND: The simultaneous accumulation of different misfolded proteins in the central nervous system is a common feature in many neurodegenerative diseases. In most cases, co-occurrence of abnormal deposited proteins is observed in different brain regions and cell populations, but, in some instances, the proteins can be found in the same cellular aggregates. Co-occurrence of tau and α-synuclein (α-syn) aggregates has been described in neurodegenerative disorders with primary deposition of α-syn, such as Parkinson's disease and dementia with Lewy bodies. Although it is known that tau and α-syn have pathological synergistic effects on their mutual fibrillization, the underlying biological effects remain unclear. METHODOLOGY/PRINCIPAL FINDINGS: We used different cell models of synucleinopathy to investigate the effects of tau on α-syn aggregation. Using confocal microscopy and FRET-based techniques we observed that tau colocalized and interacted with α-syn aggregates. We also found that tau overexpression changed the pattern of α-syn aggregation, reducing the size and increasing the number of aggregates. This shift was accompanied by an increase in the levels of insoluble α-syn. Furthermore, co-transfection of tau increased secreted α-syn and cytotoxicity. CONCLUSIONS/SIGNIFICANCE: Our data suggest that tau enhances α-syn aggregation and toxicity and disrupts α-syn inclusion formation. This pathological synergistic effect between tau and α-syn may amplify the deleterious process and spread the damage in neurodegenerative diseases that show co-occurrence of both pathologies.


Subject(s)
Models, Biological , alpha-Synuclein/metabolism , tau Proteins/physiology , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Fluorescence Resonance Energy Transfer , Humans , Microscopy, Confocal , Molecular Weight , alpha-Synuclein/biosynthesis , alpha-Synuclein/toxicity
9.
CNS Neurol Disord Drug Targets ; 9(6): 727-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20942789

ABSTRACT

Tauopathies are neurodegenerative diseases characterized by insoluble hyperphosphorylated deposits of the microtubule-associated protein tau in the central nervous system. In these disorders, tau is believed to cause neurodegeneration and neuronal loss due to the loss of function of the normal protein, and/or the gain of toxic properties by generating multimeric species. The obstacles found in amyloid-based therapies in Alzheimer's disease, the most common tauopathy, have stimulated the search for alternative targets, including tau. In this article, we review the strategies aimed at reducing tau phosphorylation and aggregation as a target for drug intervention in tauopathies.


Subject(s)
Drug Delivery Systems/methods , Drugs, Investigational/therapeutic use , Tauopathies/drug therapy , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Humans , Models, Neurological , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurofibrillary Tangles/pathology , Phosphorylation , Tauopathies/genetics , Tauopathies/pathology , tau Proteins/genetics
10.
Mol Biol Cell ; 20(24): 5051-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19846661

ABSTRACT

Bone morphogenetic proteins (BMPs) have been implicated in the generation and postnatal differentiation of cerebellar granule cells (CGCs). Here, we examined the eventual role of BMPs on the survival of these neurons. Lack of depolarization causes CGC death by apoptosis in vivo, a phenomenon that is mimicked in vitro by deprivation of high potassium in cultured CGCs. We have found that BMP-6, but not BMP-7, is able to block low potassium-mediated apoptosis in CGCs. The neuroprotective effect of BMP-6 is not accompanied by an increase of Smad translocation to the nucleus, suggesting that the canonical pathway is not involved. By contrast, activation of the MEK/ERK/CREB pathway by BMP-6 is necessary for its neuroprotective effect, which involves inhibition of caspase activity and an increase in Bcl-2 protein levels. Other pathways involved in the regulation of CGC survival, such as the c-Jun terminal kinase and the phosphatidylinositol 3-kinase (PI3K)-Akt/PKB, were not affected by BMP-6. Moreover, failure of BMP-7 to activate the MEK/ERK/CREB pathway could explain its inability to protect CGCs from low potassium-mediated apoptosis. Thus, this study demonstrates that BMP-6 acting through the noncanonical MEK/ERK/CREB pathway plays a crucial role on CGC survival.


Subject(s)
Bone Morphogenetic Protein 6/pharmacology , Cerebellum/cytology , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/cytology , Animals , Apoptosis/drug effects , Caspase Inhibitors , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cytoprotection/drug effects , Enzyme Activation/drug effects , Models, Biological , Neurons/enzymology , Potassium/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Smad Proteins/metabolism
11.
Neurobiol Dis ; 35(3): 438-47, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19555759

ABSTRACT

TNF-alpha has been reported to be relevant in stroke-induced neuronal death. However the precise function of TNF-alpha in brain ischemia remains controversial since there are data supporting either a detrimental or a protective effect. Here we show that TNF-alpha is released after oxygen-glucose deprivation (OGD) of cortical cultures and is a major contributor to the apoptotic death observed without affecting the OGD-mediated necrotic cell death. In this paradigm, apoptosis depends on TNF-alpha-induced activation of caspase-8 and -3 without affecting the activation of caspase-9. By using knock-out mice for TNF-alpha receptor 1, we show that the activation of both caspase-3 and -8 by TNF-alpha is mediated by TNF-alpha receptor 1. The pro-apoptotic role of TNF-alpha in OGD is restricted to neurons and microglia, since astrocytes do not express either TNF-alpha or TNF-alpha receptor 1. Altogether, these results show that apoptosis of cortical neurons after OGD is mediated by TNF-alpha/TNF-alpha receptor 1.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , Caspase 8/metabolism , Cerebral Cortex/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/cytology , Glucose/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/physiology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/genetics
12.
Cell Res ; 18(10): 1020-36, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18591962

ABSTRACT

Upon activation, tumor necrosis factor alpha (TNF-alpha) receptor can engage apoptotic or survival pathways. Inhibition of macromolecular synthesis is known to sensitize cells to TNF-alpha-induced cell death. It is believed that this sensitization is due to the transcriptional blockade of genes regulated by NF-kappaB. Nevertheless, such evidence has remained elusive in the nervous system. Here, we show that TNF-alpha cannot normally induce apoptosis in PC12 cells or cortical neurons. However, cells treated with Actinomycin D (ActD) become susceptible to TNF-alpha-induced cell death through the activation of caspase-8, generation of tBid and activation of caspase-9 and -3. Analysis of several proteins involved in TNF-alpha receptor signaling showed no significant downregulation of NF-kappaB target genes, such as IAPs or FLIP, under such conditions. However, Bcl-x(L) protein levels, but not those of Bcl-2, Bax and Bak, are reduced by ActD or TNF-alpha/ActD treatments. Moreover, Bcl-x(L) overexpression fully protects cells against TNF-alpha/ActD-induced cell death. When endogenous levels of Bcl-x(L) are specifically downregulated by lentiviral-based RNAi, cells no longer require ActD to be sensitive to TNF-alpha-triggered apoptosis. Furthermore, Bcl-x(L) downregulation does not affect TNF-alpha-mediated NF-kappaB activation. Altogether, our results demonstrate that Bcl-x(L), and not Bcl-2, FLIP or IAPs, acts as the endogenous regulator of neuronal resistance/sensitivity to TNF-alpha-induced apoptosis in an NF-kappaB-independent manner.


Subject(s)
Apoptosis , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/toxicity , bcl-X Protein/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cells, Cultured , Dactinomycin/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , NF-kappa B/antagonists & inhibitors , PC12 Cells , Protein Synthesis Inhibitors/pharmacology , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , bcl-X Protein/genetics
13.
J Neurosci ; 27(42): 11228-41, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17942717

ABSTRACT

Death receptors (DRs) and their ligands are expressed in developing nervous system. However, neurons are generally resistant to death induction through DRs and rather their activation promotes neuronal outgrowth and branching. These results suppose the existence of DRs antagonists expressed in the nervous system. Fas apoptosis inhibitory molecule (FAIM(S)) was first identified as a Fas antagonist in B-cells. Soon after, a longer alternative spliced isoform with unknown function was identified and named FAIM(L). FAIM(S) is widely expressed, including the nervous system, and we have shown previously that it promotes neuronal differentiation but it is not an anti-apoptotic molecule in this system. Here, we demonstrate that FAIM(L) is expressed specifically in neurons, and its expression is regulated during the development. Expression could be induced by NGF through the extracellular regulated kinase pathway in PC12 (pheochromocytoma cell line) cells. Contrary to FAIM(S), FAIM(L) does not increase the neurite outgrowth induced by neurotrophins and does not interfere with nuclear factor kappaB pathway activation as FAIM(S) does. Cells overexpressing FAIM(L) are resistant to apoptotic cell death induced by DRs such as Fas or tumor necrosis factor R1. Reduction of endogenous expression by small interfering RNA shows that endogenous FAIM(L) protects primary neurons from DR-induced cell death. The detailed analysis of this antagonism shows that FAIM(L) can bind to Fas receptor and prevent the activation of the initiator caspase-8 induced by Fas. In conclusion, our results indicate that FAIM(L) could be responsible for maintaining initiator caspases inactive after receptor engagement protecting neurons from the cytotoxic action of death ligands.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Apoptosis/physiology , Inhibitor of Apoptosis Proteins/physiology , Neurons/metabolism , Receptors, Death Domain/antagonists & inhibitors , Receptors, Death Domain/physiology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/physiology , Cells, Cultured , Gene Expression Regulation/physiology , Genetic Variation/physiology , Humans , Mice , Neurons/pathology , PC12 Cells , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Rats , Receptors, Death Domain/genetics
14.
Stroke ; 35(10): 2396-401, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15322300

ABSTRACT

BACKGROUND AND PURPOSE: Administration of histamine receptor antagonists has been reported to produce contradictory results, either reducing or increasing neural damage induced by ischemia. In this study, we investigated the neuroprotective effects of histamine H2-receptor antagonists in an "in vitro" model of ischemia. METHODS: Cultured rat brain cortical neurons were exposed to oxygen-glucose deprivation (OGD) in the presence or absence of different histaminergic drugs. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide reduction assay. Necrosis and apoptosis were quantified by staining cells with propidium iodide and Hoechst 33258. Caspase 3 activation was determined by immunocytochemistry and Western blot. RESULTS: Pretreatment with H2 antagonists effectively reduced neuronal cell death induced by OGD. Ranitidine decreased the number of necrotic and apoptotic cells. Caspase 3 activation and alteration of the neuronal cytoskeleton were also prevented by ranitidine pretreatment. The neuroprotective effect of ranitidine was still evident when added 6 hours after OGD. CONCLUSIONS: H2-receptor antagonists protected against OGD-induced neuronal death. Ranitidine attenuated cell death even when administered after OGD. These data suggest that this drug, which is currently used for the treatment of gastric ulcers, may be useful in promoting recovery after ischemia.


Subject(s)
Brain Ischemia/pathology , Cell Death/drug effects , Histamine H2 Antagonists/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Ranitidine/pharmacology , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Survival , Cells, Cultured , Glucose , Necrosis , Oxygen , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...