Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 10(1): 49, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272891

ABSTRACT

Electrical pulse stimulation has been used to enhance the differentiation or proliferation of neuronal progenitor cells in tissue engineering and cancer treatment. Therefore, a comprehensive investigation of the effects caused by its parameters is crucial for improvements in those fields. We propose a study of pulse parameters, to allow the control of N2a cell line fate and behavior. We have focused on designing an experimental setup that allows for the knowledge and control over the environment and the stimulation signals applied. To map the effects of the stimulation on N2a cells, their morphology and the cellular and molecular reactions induced by the pulse stimulation have been analyzed. Immunofluorescence, rt-PCR and western blot analysis have been carried out for this purpose, as well as cell counting. Our results show that low-amplitude electrical pulse stimulation promotes proliferation of N2a cells, whilst amplitudes in the range 250 mV/mm-500 mV/mm induce differentiation. Amplitudes higher than 750 mV/mm produce cell damage at low frequencies. For high frequencies, large amplitudes are needed to cause cell death. An inverse relation has been found between cell density and pulse-induced neuronal differentiation. The best condition for neuronal differentiation was found to be 500 mV/mm at 100 Hz. These findings have been confirmed by up-regulation of the Neurod1 gene. Our preliminary study of the molecular effects of electrical pulse stimulation on N2a offers premonitory clues of the PI3K/Akt/GSK-3ß pathway implications on the neuronal differentiation process through ES. In general, we have successfully mapped the sensitivity of N2a cells to electrical pulse stimulation parameters.

2.
Cells ; 13(2)2024 01 09.
Article in English | MEDLINE | ID: mdl-38247815

ABSTRACT

The COVID-19 pandemic has brought to the forefront the intricate relationship between SARS-CoV-2 and its impact on neurological complications, including potential links to neurodegenerative processes, characterized by a dysfunction of the protein quality control systems and ER stress. This review article explores the role of protein quality control systems, such as the Unfolded Protein Response (UPR), the Endoplasmic Reticulum-Associated Degradation (ERAD), the Ubiquitin-Proteasome System (UPS), autophagy and the molecular chaperones, in SARS-CoV-2 infection. Our hypothesis suggests that SARS-CoV-2 produces ER stress and exploits the protein quality control systems, leading to a disruption in proteostasis that cannot be solved by the host cell. This disruption culminates in cell death and may represent a link between SARS-CoV-2 and neurodegeneration.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/complications , Endoplasmic Reticulum-Associated Degradation , Pandemics , Ubiquitin
3.
Front Mol Biosci ; 8: 658742, 2021.
Article in English | MEDLINE | ID: mdl-34222330

ABSTRACT

Aging is a biological and multifactorial process characterized by a progressive and irreversible deterioration of the physiological functions leading to a progressive increase in morbidity. In the next decades, the world population is expected to reach ten billion, and globally, elderly people over 80 are projected to triple in 2050. Consequently, it is also expected an increase in the incidence of age-related pathologies such as cancer, diabetes, or neurodegenerative disorders. Disturbance of cellular protein homeostasis (proteostasis) is a hallmark of normal aging that increases cell vulnerability and might be involved in the etiology of several age-related diseases. This review will focus on the molecular alterations occurring during normal aging in the most relevant protein quality control systems such as molecular chaperones, the UPS, and the ALS. Also, alterations in their functional cooperation will be analyzed. Finally, the role of inflammation, as a synergistic negative factor of the protein quality control systems during normal aging, will also be addressed. A better comprehension of the age-dependent modifications affecting the cellular proteostasis, as well as the knowledge of the mechanisms underlying these alterations, might be very helpful to identify relevant risk factors that could be responsible for or contribute to cell deterioration, a fundamental question still pending in biomedicine.

4.
Aging Cell ; 19(11): e13257, 2020 11.
Article in English | MEDLINE | ID: mdl-33146912

ABSTRACT

Diabetes and metabolic syndrome are associated with the typical American high glycemia diet and result in accumulation of high levels of advanced glycation end products (AGEs), particularly upon aging. AGEs form when sugars or their metabolites react with proteins. Associated with a myriad of age-related diseases, AGEs accumulate in many tissues and are cytotoxic. To date, efforts to limit glycation pharmacologically have failed in human trials. Thus, it is crucial to identify systems that remove AGEs, but such research is scanty. Here, we determined if and how AGEs might be cleared by autophagy. Our in vivo mouse and C. elegans models, in which we altered proteolysis or glycative burden, as well as experiments in five types of cells, revealed more than six criteria indicating that p62-dependent autophagy is a conserved pathway that plays a critical role in the removal of AGEs. Activation of autophagic removal of AGEs requires p62, and blocking this pathway results in accumulation of AGEs and compromised viability. Deficiency of p62 accelerates accumulation of AGEs in soluble and insoluble fractions. p62 itself is subject to glycative inactivation and accumulates as high mass species. Accumulation of p62 in retinal pigment epithelium is reversed by switching to a lower glycemia diet. Since diminution of glycative damage is associated with reduced risk for age-related diseases, including age-related macular degeneration, cardiovascular disease, diabetes, Alzheimer's, and Parkinson's, discovery of methods to limit AGEs or enhance p62-dependent autophagy offers novel potential therapeutic targets to treat AGEs-related pathologies.


Subject(s)
Glycation End Products, Advanced/metabolism , RNA-Binding Proteins/metabolism , Animals , Autophagy/physiology , Cell Line , Cell Survival/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Lysosomes , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats
5.
Cell Death Dis ; 9(5): 531, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29748539

ABSTRACT

Sirtuin 1 (SIRT1) activity is neuroprotective, and we have recently demonstrated its role in the retrograde degenerative process in motoneurons (MNs) in the spinal cord of rats after peripheral nerve root avulsion (RA) injury. SIRT2 has been suggested to exert effects opposite those of SIRT1; however, its roles in neurodegeneration and neuron response after nerve injury remain unclear. Here we compared the neuroprotective potentials of SIRT1 activation and SIRT2 inhibition in a mouse model of hypoglossal nerve axotomy. This injury induced a reduction of around half MN population within the hypoglossal nucleus by a non-apoptotic neurodegenerative process triggered by endoplasmic reticulum (ER) stress that resulted in activation of the unfolded protein response mediated by IRE1α and XBP1 by 21 days post injury. Both SIRT1 activation with NeuroHeal and SIRT2 inhibition with AK7 protected NSC-34 motor neuron-like cells against ER stress in vitro. In agreement with the in vitro results, NeuroHeal treatment or SIRT1 overexpression was neuroprotective of axotomized hypoglossal MNs in a transgenic mouse model. In contrast, AK7 treatment or SIRT2 genetic depletion in mice inhibited damaged MN survival. To resolve the in vitro/in vivo discrepancies, we used an organotypic spinal cord culture system that preserves glial cells. In this system, AK7 treatment of ER-stressed organotypic cultures was detrimental for MNs and increased microglial nuclear factor-κB and the consequent transcription of cytotoxic pro-inflammatory factors similarly. The results highlight the importance of glial cells in determining the neuroprotective impact of any treatment.


Subject(s)
Acamprosate/pharmacology , Benzamides/pharmacology , Hypoglossal Nerve Injuries , Motor Neurons/enzymology , Neuroprotection/drug effects , Ribavirin/pharmacology , Sirtuin 1 , Sirtuin 2 , Sulfonamides/pharmacology , Animals , Drug Combinations , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Female , Hypoglossal Nerve Injuries/drug therapy , Hypoglossal Nerve Injuries/enzymology , Hypoglossal Nerve Injuries/genetics , Hypoglossal Nerve Injuries/pathology , Mice , Mice, Knockout , Motor Neurons/pathology , Neuroprotection/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/genetics , Sirtuin 2/metabolism
6.
Sci Rep ; 7(1): 8100, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28808322

ABSTRACT

Proteostasis alteration and neuroinflammation are typical features of normal aging. We have previously shown that neuroinflammation alters cellular proteostasis through immunoproteasome induction, leading to a transient decrease of proteasome activity. Here, we further investigated the role of acute lipopolysaccharide (LPS)-induced hippocampal neuroinflammation in cellular proteostasis. In particular, we focused on macroautophagy (hereinafter called autophagy) and endoplasmic reticulum-associated protein degradation (ERAD). We demonstrate that LPS injection induced autophagy activation that was dependent, at least in part, on glycogen synthase kinase (GSK)-3ß activity but independent of mammalian target of rapamycin (mTOR) inhibition. Neuroinflammation also produced endoplasmic reticulum (ER) stress leading to canonical unfolded protein response (UPR) activation with a rapid activating transcription factor (ATF) 6α attenuation that resulted in a time-dependent down-regulation of ERAD markers. In this regard, the time-dependent accumulation of unspliced X-box binding protein (XBP) 1, likely because of decreased inositol-requiring enzyme (IRE) 1α-mediated splicing activity, might underlie in vivo ATF6α attenuation. Importantly, lactacystin-induced activation of ERAD was abolished in both the acute neuroinflammation model and in aged rats. Therefore, we provide a cellular pathway through which neuroinflammation might sensitize cells to neurodegeneration under stress situations, being relevant in normal aging and other disorders where neuroinflammation is a characteristic feature.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum-Associated Degradation/physiology , Inflammation/physiopathology , Proteostasis/physiology , Activating Transcription Factor 6/metabolism , Animals , Cell Line , Down-Regulation/physiology , Endoribonucleases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation/metabolism , Male , Mice , Rats , Rats, Wistar , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , X-Box Binding Protein 1/metabolism
7.
Sci Rep ; 5: 10027, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25941117

ABSTRACT

Targeting the ubiquitin proteasome pathway has emerged as a rational approach in the treatment of human cancers. Autophagy has been described as a cytoprotective mechanism to increase tumor cell survival under stress conditions. Here, we have focused on the role of proteasome inhibition in cell cycle progression and the role of autophagy in the proliferation recovery. The study was performed in the breast cancer cell line MCF7 compared to the normal mammary cell line MCF10A. We found that the proteasome inhibitor MG132 induced G1/S arrest in MCF10A, but G2/M arrest in MCF7 cells. The effect of MG132 on MCF7 was reproduced on MCF10A cells in the presence of the glycogen synthase kinase 3ß (GSK-3ß) inhibitor VII. Similarly, MCF7 cells overexpressing constitutively active GSK-3ß behaved like MCF10A cells. On the other hand, MCF10A cells remained arrested after MG132 removal while MCF7 recovered the proliferative capacity. Importantly, this recovery was abolished in the presence of the autophagy inhibitor 3-methyladenine (3-MA). Thus, our results support the relevance of GSK-3ß and autophagy as two targets for controlling cell cycle progression and proliferative capacity in MCF7, highlighting the co-treatment of breast cancer cells with 3-MA to synergize the effect of the proteasome inhibition.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , G1 Phase Cell Cycle Checkpoints , Glycogen Synthase Kinase 3/metabolism , Proteasome Endopeptidase Complex/metabolism , S Phase Cell Cycle Checkpoints , Autophagy , Cell Proliferation , Glycogen Synthase Kinase 3 beta , Humans , MCF-7 Cells , Signal Transduction
8.
Neurobiol Aging ; 36(5): 1953-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25817083

ABSTRACT

Autophagy plays a key role in the maintenance of cellular homeostasis, and autophagy deregulation gives rise to severe disorders. Many of the signaling pathways regulating autophagy under stress conditions are still poorly understood. Using a model of proteasome stress in rat hippocampus, we have characterized the functional crosstalk between the ubiquitin proteasome system and the autophagy-lysosome pathway, identifying also age-related modifications in the crosstalk between both proteolytic systems. Under proteasome inhibition, both autophagy activation and resolution were efficiently induced in young but not in aged rats, leading to restoration of protein homeostasis only in young pyramidal neurons. Importantly, proteasome stress inhibited glycogen synthase kinase-3ß in young but activated in aged rats. This age-related difference could be because of a dysfunction in the signaling pathway of the insulin growth factor-1 under stress situations. Present data highlight the potential role of glycogen synthase kinase-3ß in the coordination of both proteolytic systems under stress situation, representing a key molecular target to sort out this deleterious effect.


Subject(s)
Aging/metabolism , Aging/physiology , Autophagy/physiology , Glycogen Synthase Kinase 3/physiology , Hippocampus/physiology , Lysosomes/physiology , Proteasome Endopeptidase Complex/physiology , Pyramidal Cells/metabolism , Signal Transduction/physiology , Animals , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Homeostasis , Insulin-Like Growth Factor I/metabolism , Male , Neurodegenerative Diseases/genetics , Proteasome Inhibitors , Proteins/metabolism , Proteolysis , Pyramidal Cells/physiology , Rats, Wistar , Ubiquitin/physiology
9.
Front Mol Neurosci ; 6: 54, 2014.
Article in English | MEDLINE | ID: mdl-24427113

ABSTRACT

PI3K activation promotes the formation of synaptic contacts and dendritic spines, morphological features of glutamatergic synapses that are commonly known to be related to learning processes. In this report, we show that in vivo administration of a peptide that activates the PI3K signaling pathway increases spine density in the rat hippocampus and enhances the animals' cognitive abilities, while in vivo electrophysiological recordings show that PI3K activation results in synaptic enhancement of Schaffer and stratum lacunosum moleculare inputs. Morphological characterization of the spines reveals that subjecting the animals to contextual fear-conditioning training per se promotes the formation of large spines, while PI3K activation reverts this effect and favors a general change toward small head areas. Studies using hippocampal neuronal cultures show that the PI3K spinogenic process is NMDA-dependent and activity-independent. In culture, PI3K activation was followed by mRNA upregulation of glutamate receptor subunits and of the immediate-early gene Arc. Time-lapse studies confirmed the ability of PI3K to induce the formation of small spines. Finally, we demonstrate that the spinogenic effect of PI3K can be induced in the presence of neurodegeneration, such as in the Tg2576 Alzheimer's mouse model. These findings highlight that the PI3K pathway is an important regulator of neuronal connectivity and stress the relationship between spine size and learning processes.

10.
J Agric Food Chem ; 60(50): 12304-11, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23176387

ABSTRACT

Neuroinflammation is an important contributor to pathogenesis of age-related neurodegenerative disorders such as Alzheimer's or Parkinson's disease. Accumulating evidence indicates that inhibition of microglia-mediated neuroinflammation may become a reliable protective strategy for neurodegenerative processes. Flavonoids, widely distributed in the vegetable kingdom and in foods such as honey, have been suggested as novel therapeutic agents for the reduction of the deleterious effects of neuroinflammation. The present study investigated the potential protective effect of a honey flavonoid extract (HFE) on the production of pro-inflammatory mediators by lipopolysaccharide-stimulated N13 microglia. The results show that HFE significantly inhibited the release of pro-inflammatory cytokines such as TNF-α and IL-1ß. The expressions of iNOS and the production of reactive oxygen intermediates (ROS) were also significantly inhibited. Accordingly, the present study demonstrates that HFE is a potent inhibitor of microglial activation and thus a potential preventive-therapeutic agent for neurodegenerative diseases involving neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Honey/analysis , Lipopolysaccharides/pharmacology , Microglia/drug effects , Animals , Cell Line , Chromatography, High Pressure Liquid , Cytokines/biosynthesis , Cytokines/metabolism , Flavonoids/isolation & purification , Mice , Microglia/metabolism , Reactive Oxygen Species/metabolism , Tandem Mass Spectrometry
11.
J Neurochem ; 123(4): 635-44, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22913583

ABSTRACT

Regulation of proteasome abundance to meet cell needs under stress conditions is critical for maintaining cellular homeostasis. However, the effects of aging on this homeostatic response remain unknown. In this report, we analyzed in young and aged rat hippocampus, the dynamics of proteasome recovery induced by proteasome stress. Proteasome inhibition in young rats leads to an early and coordinate transcriptional and translational up-regulation of both the catalytic subunits of constitutive proteasome and the proteasome maturation protein. By contrast, aged rats up-regulated the inducible catalytic subunits and showed a lower and shorter expression of proteasome maturation protein. This resulted in a faster recovery of proteasome activity in young rats. Importantly, proteasome inhibition highly affected pyramidal cells, leading to the accumulation of ubiquitinated proteins in perinuclear regions of aged, but not young pyramidal neurons. These data strongly suggest that age-dependent differences in proteasome level and composition could contribute to neurodegeneration induced by proteasome dysfunction in normal and pathological aging.


Subject(s)
Aging , Hippocampus/metabolism , Proteasome Endopeptidase Complex/metabolism , Up-Regulation/physiology , Age Factors , Animals , Catalytic Domain/physiology , Cell Nucleolus/metabolism , Hippocampus/cytology , Immunoproteins/metabolism , Male , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar
12.
J Neuroinflammation ; 9: 87, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22559833

ABSTRACT

BACKGROUND: Neuroinflammation and protein accumulation are characteristic hallmarks of both normal aging and age-related neurodegenerative diseases. However, the relationship between these factors in neurodegenerative processes is poorly understood. We have previously shown that proteasome inhibition produced higher neurodegeneration in aged than in young rats, suggesting that other additional age-related events could be involved in neurodegeneration. We evaluated the role of lipopolysaccharide (LPS)-induced neuroinflammation as a potential synergic risk factor for hippocampal neurodegeneration induced by proteasome inhibition. METHODS: Young male Wistar rats were injected with 1 µL of saline or LPS (5 mg/mL) into the hippocampus to evaluate the effect of LPS-induced neuroinflammation on protein homeostasis. The synergic effect of LPS and proteasome inhibition was analyzed in young rats that first received 1 µL of LPS and 24 h later 1 µL (5 mg/mL) of the proteasome inhibitor lactacystin. Animals were sacrificed at different times post-injection and hippocampi isolated and processed for gene expression analysis by real-time polymerase chain reaction; protein expression analysis by western blots; proteasome activity by fluorescence spectroscopy; immunofluorescence analysis by confocal microscopy; and degeneration assay by Fluoro-Jade B staining. RESULTS: LPS injection produced the accumulation of ubiquitinated proteins in hippocampal neurons, increased expression of the E2 ubiquitin-conjugating enzyme UB2L6, decreased proteasome activity and increased immunoproteasome content. However, LPS injection was not sufficient to produce neurodegeneration. The combination of neuroinflammation and proteasome inhibition leads to higher neuronal accumulation of ubiquitinated proteins, predominant expression of pro-apoptotic markers and increased neurodegeneration, when compared with LPS or lactacystin (LT) injection alone. CONCLUSIONS: Our results identify neuroinflammation as a risk factor that increases susceptibility to neurodegeneration induced by proteasome inhibition. These results highlight the modulation of neuroinflammation as a mechanism for neuronal protection that could be relevant in situations where both factors are present, such as aging and neurodegenerative diseases.


Subject(s)
Hippocampus/drug effects , Lipopolysaccharides/toxicity , Nerve Degeneration/chemically induced , Proteasome Inhibitors/toxicity , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Aging/drug effects , Aging/physiology , Animals , Drug Synergism , Hippocampus/enzymology , Hippocampus/pathology , Inflammation/chemically induced , Inflammation/epidemiology , Inflammation/pathology , Male , Nerve Degeneration/epidemiology , Nerve Degeneration/pathology , Proteasome Endopeptidase Complex/metabolism , Rats , Rats, Wistar , Ubiquitination/drug effects , Ubiquitination/physiology
13.
Neurobiol Aging ; 33(10): 2462-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22177721

ABSTRACT

Lewy bodies containing the centrosomal protein γ-tubulin and fragmentation of Golgi apparatus (GA) have been described in nigral neurons of Parkinson's disease (PD) patients. However, the relevance of these features in PD pathophysiology remains unknown. We analyzed the impact of proteasome inhibition in the formation of γ-tubulin-containing aggregates as well as on GA structure. SH-SY5Y cells were treated with the proteasome inhibitor Z-Leu-Leu-Leu-al (MG132) to induce centrosomal-protein aggregates. Then, microtubules (MTs) and Golgi dynamics, as well as the vesicular transport of dopamine transporter (DAT) were evaluated both in vitro and in living cells. MG132 treatment induced γ-tubulin aggregates which altered microtubule nucleation. MG132-treated cells containing γ-tubulin aggregates showed fragmentation of GA and perturbation of the trans-Golgi network. Under these conditions, the DAT accumulated at the centrosomal-Golgi region indicating that the vesicular transport of DAT was disrupted. Thus, centrosomal aggregates and fragmentation of GA are 2 closely related processes that could result in the disruption of the vesicular transport of DAT toward the plasma membrane in a model of dopaminergic neuronal degeneration.


Subject(s)
Centrosome/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Golgi Apparatus/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Cell Line , Centrosome/drug effects , Golgi Apparatus/drug effects , Humans , Leupeptins/pharmacology , Microtubules/metabolism , Proteasome Inhibitors/pharmacology , Tubulin/metabolism
14.
Acta Neuropathol ; 123(1): 53-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22020633

ABSTRACT

Dystrophic neurites associated with amyloid plaques precede neuronal death and manifest early in Alzheimer's disease (AD). In this work we have characterized the plaque-associated neuritic pathology in the hippocampus of young (4- to 6-month-old) PS1(M146L)/APP(751SL) mice model, as the initial degenerative process underlying functional disturbance prior to neuronal loss. Neuritic plaques accounted for almost all fibrillar deposits and an axonal origin of the dystrophies was demonstrated. The early induction of autophagy pathology was evidenced by increased protein levels of the autophagosome marker LC3 that was localized in the axonal dystrophies, and by electron microscopic identification of numerous autophagic vesicles filling and causing the axonal swellings. Early neuritic cytoskeletal defects determined by the presence of phosphorylated tau (AT8-positive) and actin-cofilin rods along with decreased levels of kinesin-1 and dynein motor proteins could be responsible for this extensive vesicle accumulation within dystrophic neurites. Although microsomal Aß oligomers were identified, the presence of A11-immunopositive Aß plaques also suggested a direct role of plaque-associated Aß oligomers in defective axonal transport and disease progression. Most importantly, presynaptic terminals morphologically disrupted by abnormal autophagic vesicle buildup were identified ultrastructurally and further supported by synaptosome isolation. Finally, these early abnormalities in axonal and presynaptic structures might represent the morphological substrate of hippocampal dysfunction preceding synaptic and neuronal loss and could significantly contribute to AD pathology in the preclinical stages.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Axons/ultrastructure , Cytoplasmic Vesicles/ultrastructure , Hippocampus/ultrastructure , Neurites/ultrastructure , Plaque, Amyloid/ultrastructure , Alzheimer Disease/metabolism , Animals , Autophagy , Disease Models, Animal , Hippocampus/metabolism , Mice , Mice, Transgenic , Microscopy, Immunoelectron , Neurons/metabolism , Plaque, Amyloid/metabolism , Presynaptic Terminals/ultrastructure
15.
J Neuroimmunol ; 238(1-2): 44-51, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21803430

ABSTRACT

To elucidate whether density of cells could contribute to the extent of microglial activation, we performed in vitro assays using three different densities of N13 microglia stimulated with LPS. Our results showed that induction of pro-inflammatory factors as TNF-α and iNOS was directly related to cell density, meanwhile the induction of the anti-inflammatory IL-10 was inversely related to cell density. Accordingly, in vivo assays showed that after LPS-injection, iNOS expression was more intense in substantia nigra, a brain area showing specific susceptibility to neurodegeneration after microglia activation, whereas IL-10 expression was more sustained in striatum, an area resistant to damage. These results support that microglia density is pivotal to control the balance between pro- and anti-inflammatory factors release.


Subject(s)
Cytokines/metabolism , Encephalitis/chemically induced , Encephalitis/pathology , Lipopolysaccharides/toxicity , Microglia/drug effects , Nitric Oxide Synthase Type II/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Count , Cell Line, Transformed , Cytokines/genetics , Disease Models, Animal , Encephalitis/drug therapy , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation , Imidazoles/therapeutic use , Interleukin-10/genetics , Interleukin-10/metabolism , Isothiuronium/analogs & derivatives , Isothiuronium/therapeutic use , Male , Mice , Microglia/pathology , Nitric Oxide Synthase Type II/genetics , Pyridines/therapeutic use , RNA, Messenger/metabolism , Rats , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
J Biol Chem ; 286(21): 18414-25, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21460223

ABSTRACT

Neurotrophins, activating the PI3K/Akt signaling pathway, control neuronal survival and plasticity. Alterations in NGF, BDNF, IGF-1, or insulin signaling are implicated in the pathogenesis of Alzheimer disease. We have previously characterized a bigenic PS1×APP transgenic mouse displaying early hippocampal Aß deposition (3 to 4 months) but late (17 to 18 months) neurodegeneration of pyramidal cells, paralleled to the accumulation of soluble Aß oligomers. We hypothesized that PI3K/Akt/GSK-3ß signaling pathway could be involved in this apparent age-dependent neuroprotective/neurodegenerative status. In fact, our data demonstrated that, as compared with age-matched nontransgenic controls, the Ser-9 phosphorylation of GSK-3ß was increased in the 6-month PS1×APP hippocampus, whereas in aged PS1×APP animals (18 months), GSK-3ß phosphorylation levels displayed a marked decrease. Using N2a and primary neuronal cell cultures, we demonstrated that soluble amyloid precursor protein-α (sAPPα), the predominant APP-derived fragment in young PS1×APP mice, acting through IGF-1 and/or insulin receptors, activated the PI3K/Akt pathway, phosphorylated the GSK-3ß activity, and in consequence, exerted a neuroprotective action. On the contrary, several oligomeric Aß forms, present in the soluble fractions of aged PS1×APP mice, inhibited the induced phosphorylation of Akt/GSK-3ß and decreased the neuronal survival. Furthermore, synthetic Aß oligomers blocked the effect mediated by different neurotrophins (NGF, BDNF, insulin, and IGF-1) and sAPPα, displaying high selectivity for NGF. In conclusion, the age-dependent appearance of APP-derived soluble factors modulated the PI3K/Akt/GSK-3ß signaling pathway through the major neurotrophin receptors. sAPPα stimulated and Aß oligomers blocked the prosurvival signaling. Our data might provide insights into the selective vulnerability of specific neuronal groups in Alzheimer disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Glycogen Synthase Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Aging/genetics , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Cell Survival/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurons/metabolism , Neurons/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin
17.
J Alzheimers Dis ; 21(1): 119-32, 2010.
Article in English | MEDLINE | ID: mdl-20413859

ABSTRACT

Specific neuronal networks are preferentially affected in the early stages of Alzheimer's disease (AD). The distinct subpopulations of hippocampal inhibitory GABAergic system have been shown to display differential vulnerability to neurodegeneration in AD. We have previously reported a substantial loss of SOM/NPY interneurons, whereas those expressing parvalbumin were unaltered, in the hippocampus of 6 month-old PS1/AbetaPP transgenic mice. In the present study, we now investigated the pathological changes of hippocampal calretinin (CR) interneurons in this PS1/AbetaPP model from 2 to 12 months of age. The total number of CR-immunoreactive inhibitory cells was determined by stereology in CA1 and CA2/3 subfields. Our findings show a substantial decrease (35%-45%) of CR-positive interneurons in both hippocampal subfields of PS1/AbetaPP mice at very early age (4 months) compared to age-matched control mice. This decrease was accompanied by a reduced CR mRNA content as determined by quantitative RT-PCR. However, the number of another hippocampal CR-positive population (belonging to Cajal-Retzius cells) was not affected. The selective early loss of CR-interneurons was parallel to the appearance of extracellular Abeta deposits, preferentially in CR-axonal fields, and the formation of dystrophic neurites. This specific GABAergic subpopulation plays a crucial role in the generation of synchronous rhythmic activity in hippocampus by controlling other interneurons. Therefore, early alterations of hippocampal inhibitory functionality in AD, caused by select CR-cells neurodegeneration, could result in cognitive impairments seen in initial stages of the disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Hippocampus/pathology , Interneurons/metabolism , Presenilin-1/genetics , S100 Calcium Binding Protein G/metabolism , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Analysis of Variance , Animals , Calbindin 2 , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphopyruvate Hydratase/metabolism
18.
Neurochem Int ; 56(8): 878-85, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20350575

ABSTRACT

This study describes the effect of global brain ischemia followed by 48 h reperfusion, when delayed neuronal death can be already observed. We quantified the mRNA levels of the N-methyl-D-aspartate receptor (NMDAR) subunits and those of the astroglia (glial fibrilar acidic protein, GFAP) and microglia (CD11b) markers using real time PCR on the cerebral cortex and hippocampus of 3- and 18-month-old Sprague-Dawley rats. Data show an ischemia/reperfusion-induced decrease in the mRNA levels of the NMDAR NR1, NR2A and NR2B subunits genes, which contrasts with the increase in the CD11b and GFAP mRNA levels. These effects are attenuated in all the genes studied in 18-month-old animals, suggesting that this mechanism of response is less efficient in aged animals. Western blot assays of NR1, NR2A and NR2B show parallels with the real time PCR data, indicating that the down-regulation of these genes is controlled at the transcriptional level. We suggest that a decrease in the efficiency in the control of the NMDAR transcription could account for the higher vulnerability in aged animals, but it cannot explain by itself differences in the vulnerability to ischemia in different areas of the brain. In the assays of ischemia/reperfusion followed by a treatment with the anti-inflammatory agent meloxicam, we observed that ischemic insult was unable to elicit changes in the NMDAR transcription, thus suggesting that inflammation plays a crucial role in the transcriptional control of these genes.


Subject(s)
Aging/physiology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Ischemia/metabolism , Down-Regulation/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/biosynthesis , Reperfusion Injury/metabolism , Thiazines/pharmacology , Thiazoles/pharmacology , Aging/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Disease Models, Animal , Down-Regulation/genetics , Male , Meloxicam , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Reperfusion Injury/drug therapy , Thiazines/therapeutic use , Thiazoles/therapeutic use , Treatment Outcome
19.
Aging Cell ; 8(6): 654-65, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19747230

ABSTRACT

Dysfunctions of the ubiquitin proteasome system (UPS) have been proposed to be involved in the aetiology and/or progression of several age-related neurodegenerative disorders. However, the mechanisms linking proteasome dysfunction to cell degeneration are poorly understood. We examined in young and aged rat hippocampus the activation of the unfolded protein response (UPR) under cellular stress induced by proteasome inhibition. Lactacystin injection blocked proteasome activity in young and aged animals in a similar extent and increased the amount of ubiquitinated proteins. Young animals activated the three UPR arms, IRE1alpha, ATF6alpha and PERK, whereas aged rats failed to induce the IRE1alpha and ATF6alpha pathways. In consequence, aged animals did not induce the expression of pro-survival factors (chaperones, Bcl-XL and Bcl-2), displayed a more sustained expression of pro-apoptotic markers (CHOP, Bax, Bak and JKN), an increased caspase-3 processing. At the cellular level, proteasome inhibition induced neuronal damage in young and aged animals as assayed using Fluorojade-B staining. However, degenerating neurons were evident as soon as 24 h postinjection in aged rats, but it was delayed up to 3 days in young animals. Our findings show evidence supporting age-related dysfunctions in the UPR activation as a potential mechanism linking protein accumulation to cell degeneration. An imbalance between pro-survival and pro-apoptotic proteins, because of noncanonical activation of the UPR in aged rats, would increase the susceptibility to cell degeneration. These findings add a new molecular vision that might be relevant in the aetiology of several age-related neurodegenerative disorders.


Subject(s)
Acetylcysteine/analogs & derivatives , Aging , Hippocampus/metabolism , Hippocampus/pathology , Nerve Degeneration/metabolism , Proteasome Inhibitors , Unfolded Protein Response/drug effects , Acetylcysteine/pharmacology , Animals , Biomarkers , Caspase 3/metabolism , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Male , Nerve Degeneration/chemically induced , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Signal Transduction
20.
J Alzheimers Dis ; 18(4): 755-76, 2009.
Article in English | MEDLINE | ID: mdl-19661615

ABSTRACT

Here we demonstrated that extracellular, not intracellular, amyloid-beta (Abeta) and the associated cytotoxic glial neuroinflammatory response are major contributors to early neuronal loss in a PS1xAPP model. A significant loss of principal (27%) and SOM/NPY (56-46%) neurons was found in the entorhinal cortex at 6 months of age. Loss of principal cells occurred selectively in deep layers (primarily layer V) whereas SOM/NPY cell loss was evenly distributed along the cortical column. Neither layer V pyramidal neurons nor SOM/NPY interneurons displayed intracellular Abeta immunoreactivity, even after formic acid retrieval; thus, extracellular factors should be preferentially implicated in this selective neurodegeneration. Amyloid deposits were mainly concentrated in deep layers at 4-6 months, and of relevance was the existence of a potentially cytotoxic inflammatory response (TNFalpha, TRAIL, and iNOS mRNAs were upregulated). Moreover, non-plaque associated activated microglial cells and reactive astrocytes expressed TNFalpha and iNOS, respectively. At this age, in the hippocampus of same animals, extracellular Abeta induced a non-cytotoxic glial activation. The opposite glial activation, at the same chronological age, in entorhinal cortex and hippocampus strongly support different mechanisms of disease progression in these two regions highly affected by Abeta pathology.


Subject(s)
Aging/metabolism , Amyloid beta-Peptides/metabolism , Entorhinal Cortex/pathology , Neuroglia/metabolism , Neurons/pathology , Aging/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Biomarkers/metabolism , Cell Death , Entorhinal Cortex/cytology , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , Polymerase Chain Reaction , Presenilin-1/genetics , RNA , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...