Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38474102

ABSTRACT

Histone deacetylase SIRT1 represses gene expression through the deacetylation of histones and transcription factors and is involved in the protective cell response to stress and aging. However, upon endoplasmic reticulum (ER) stress, SIRT1 impairs the IRE1α branch of the unfolded protein response (UPR) through the inhibition of the transcriptional activity of XBP-1 and SIRT1 deficiency is beneficial under these conditions. We hypothesized that SIRT1 deficiency may unlock the blockade of transcription factors unrelated to the UPR promoting the synthesis of chaperones and improving the stability of immature proteins or triggering the clearance of unfolded proteins. SIRT1+/+ and SIRT1-/- fibroblasts were exposed to the ER stress inducer tunicamycin and cell survival and expression of heat shock proteins were analyzed 24 h after the treatment. We observed that SIRT1 loss significantly reduced cell sensitivity to ER stress and showed that SIRT1-/- but not SIRT1+/+ cells constitutively expressed high levels of phospho-STAT3 and heat shock proteins. Hsp70 silencing in SIRT1-/- cells abolished the resistance to ER stress. Furthermore, accumulation of ubiquitinated proteins was lower in SIRT1-/- than in SIRT1+/+ cells. Our data showed that SIRT1 deficiency enabled chaperones upregulation and boosted the proteasome activity, two processes that are beneficial for coping with ER stress.


Subject(s)
Heat-Shock Proteins , Sirtuin 1 , Heat-Shock Proteins/metabolism , Up-Regulation , Sirtuin 1/metabolism , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum Stress , Unfolded Protein Response , Molecular Chaperones/metabolism , Transcription Factors/metabolism
2.
Open Biol ; 13(1): 220200, 2023 01.
Article in English | MEDLINE | ID: mdl-36629019

ABSTRACT

Microglia are very sensitive to changes in the environment and respond through morphological, functional and metabolic adaptations. To depict the modifications microglia undergo under healthy and pathological conditions, we developed free access image analysis scripts to quantify microglia morphologies and phagocytosis. Neuron-glia cultures, in which microglia express the reporter tdTomato, were exposed to excitotoxicity or excitotoxicity + inflammation and analysed 8 h later. Neuronal death was assessed by SYTOX staining of nucleus debris and phagocytosis was measured through the engulfment of SYTOX+ particles in microglia. We identified seven morphologies: round, hypertrophic, fried egg, bipolar and three 'inflamed' morphologies. We generated a classifier able to separate them and assign one of the seven classes to each microglia in sample images. In control cultures, round and hypertrophic morphologies were predominant. Excitotoxicity had a limited effect on the composition of the populations. By contrast, excitotoxicity + inflammation promoted an enrichment in inflamed morphologies and increased the percentage of phagocytosing microglia. Our data suggest that inflammation is critical to promote phenotypical changes in microglia. We also validated our tools for the segmentation of microglia in brain slices and performed morphometry with the obtained mask. Our method is versatile and useful to correlate microglia sub-populations and behaviour with environmental changes.


Subject(s)
Microglia , Phagocytosis , Humans , Microglia/metabolism , Inflammation/metabolism , Cell Death , Neurons/metabolism
3.
Biomedicines ; 9(8)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34440243

ABSTRACT

An experimental model of spinal root avulsion (RA) is useful to study causal molecular programs that drive retrograde neurodegeneration after neuron-target disconnection. This neurodegenerative process shares common characteristics with neuronal disease-related processes such as the presence of endoplasmic reticulum (ER) stress and autophagy flux blockage. We previously found that the overexpression of GRP78 promoted motoneuronal neuroprotection after RA. After that, we aimed to unravel the underlying mechanism by carrying out a comparative unbiased proteomic analysis and pharmacological and genetic interventions. Unexpectedly, mitochondrial factors turned out to be most altered when GRP78 was overexpressed, and the abundance of engulfed mitochondria, a hallmark of mitophagy, was also observed by electronic microscopy in RA-injured motoneurons after GRP78 overexpression. In addition, GRP78 overexpression increased LC3-mitochondria tagging, promoted PINK1 translocation, mitophagy induction, and recovered mitochondrial function in ER-stressed cells. Lastly, we found that GRP78-promoted pro-survival mitophagy was mediated by PINK1 and IP3R in our in vitro model of motoneuronal death. This data indicates a novel relationship between the GRP78 chaperone and mitophagy, opening novel therapeutical options for drug design to achieve neuroprotection.

4.
Sci Rep ; 8(1): 1879, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382857

ABSTRACT

Here we used a systems biology approach and artificial intelligence to identify a neuroprotective agent for the treatment of peripheral nerve root avulsion. Based on accumulated knowledge of the neurodegenerative and neuroprotective processes that occur in motoneurons after root avulsion, we built up protein networks and converted them into mathematical models. Unbiased proteomic data from our preclinical models were used for machine learning algorithms and for restrictions to be imposed on mathematical solutions. Solutions allowed us to identify combinations of repurposed drugs as potential neuroprotective agents and we validated them in our preclinical models. The best one, NeuroHeal, neuroprotected motoneurons, exerted anti-inflammatory properties and promoted functional locomotor recovery. NeuroHeal endorsed the activation of Sirtuin 1, which was essential for its neuroprotective effect. These results support the value of network-centric approaches for drug discovery and demonstrate the efficacy of NeuroHeal as adjuvant treatment with surgical repair for nervous system trauma.


Subject(s)
Neuroprotective Agents/pharmacology , Peripheral Nervous System Diseases/drug therapy , Wounds and Injuries/drug therapy , Algorithms , Animals , Artificial Intelligence , Cell Line , Female , Machine Learning , Mice , Nerve Regeneration/drug effects , Radiculopathy/drug therapy , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Nerve Roots/drug effects
5.
Mol Neurobiol ; 54(7): 5604-5619, 2017 09.
Article in English | MEDLINE | ID: mdl-27614878

ABSTRACT

SIRT1 induces cell survival and has shown neuroprotection against amyloid and tau pathologies in Alzheimer's disease (AD). However, protective effects against memory loss or the enhancement of cognitive functions have not yet been proven. We aimed to investigate the benefits induced by SIRT1 overexpression in the hippocampus of the AD mouse model 3xTg-AD and in control non-transgenic mice. A lentiviral vector encoding mouse SIRT1 or GFP, selectively transducing neurons, was injected into the dorsal CA1 hippocampal area of 4-month-old mice. Six-month overexpression of SIRT1 fully preserved learning and memory in 10-month-old 3xTg-AD mice. Remarkably, SIRT1 also induced cognitive enhancement in healthy non-transgenic mice. Neuron cultures of 3xTg-AD mice, which show traits of AD-like pathology, and neuron cultures from non-transgenic mice were also transduced with lentiviral vectors to analyze beneficial SIRT1 mechanisms. We uncovered novel pathways of SIRT1 neuroprotection through enhancement of cell proteostatic mechanisms and activation of neurotrophic factors not previously reported such as GDNF, present in both AD-like and healthy neurons. Therefore, SIRT1 may increase neuron function and resilience against AD.


Subject(s)
Cognition/physiology , Hippocampus/metabolism , Learning/physiology , Sirtuin 1/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Hippocampus/drug effects , Male , Mice, Transgenic , Neurons/metabolism , Nootropic Agents/metabolism
6.
Transpl Int ; 27(5): 493-503, 2014 May.
Article in English | MEDLINE | ID: mdl-24472096

ABSTRACT

Ischemia-reperfusion (IR) injury is an important problem in liver surgery especially when steatosis is present. Ischemic preconditioning (PC) is the only surgical strategy that has been applied in patients with steatotic livers undergoing warm ischemia. Silent information regulator 1 (SIRT1) is a histone deacetylase that regulates various cellular processes. This study evaluates the SIRT1 implication in PC in fatty livers. Homozygous (Ob) Zucker rats were subjected to IR and IR + PC. An additional group treated with sirtinol or EX527 (SIRT1 inhibitors) before PC was also realized. Liver injury and oxidative stress were evaluated. SIRT1 protein levels and activity, as well as other parameters involved in PC protective mechanisms (adenosine monophosphate protein kinase, eNOS, HSP70, MAP kinases, apoptosis), were also measured. We demonstrated that the protective effect of PC was due in part to SIRT1 induction, as SIRT1 inhibition resulted in increased liver injury and abolished the beneficial mechanisms of PC. In this study, we report for the first time that SIRT1 is involved in the protective mechanisms induced by hepatic PC in steatotic livers.


Subject(s)
Fatty Liver/complications , Ischemic Preconditioning , Liver/blood supply , Reperfusion Injury/prevention & control , Sirtuin 1/physiology , Animals , Apoptosis , HSP70 Heat-Shock Proteins/physiology , Liver/pathology , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type III/physiology , Oxidative Stress , Rats , Rats, Zucker , Sirtuin 1/analysis
7.
Transl Stroke Res ; 4(6): 663-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24323420

ABSTRACT

Silent information regulator 1 (SIRT1) is a NAD+-dependent histone deacetylase that represses gene expression and plays a role in longevity. SIRT1 responds to diverse stress conditions and regulates metabolism in nutrient deficiency conditions; therefore, it is involved in adaptive pathways to better fulfill tissue needs in a disturbed environment. SIRT1 overexpression or activation is protective in neurodegenerative diseases. Its role in acute nervous system injury, such as brain ischemia, is emerging, but whether SIRT1 activation improves stroke outcome is still a matter of controversy. In the present review, we will document present knowledge about the contribution of SIRT1 in death/survival in cell and animal models of brain ischemia and discuss whether SIRT1 could be a valuable target for therapeutic intervention in human stroke.


Subject(s)
Brain Ischemia/metabolism , Sirtuin 1/metabolism , Stroke/metabolism , Alzheimer Disease/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Humans , Huntington Disease/metabolism , Parkinson Disease/metabolism
8.
Biophys J ; 104(5): 968-75, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23473479

ABSTRACT

Neuronal death can be preceded by progressive dysfunction of axons. Several pathological conditions such as ischemia can disrupt the neuronal cytoskeleton. Microtubules are basic structural components of the neuronal cytoskeleton that regulate axonal transport and neuronal function. Up-to-date, high-resolution observation of microtubules in living neuronal cells is usually accomplished using fluorescent-based microscopy techniques. However, this needs exogenous fluorescence markers to produce the required contrast. This is an invasive procedure that may interfere with the microtubule dynamics. In this work, we show, for the first time to our knowledge, that by using the endogenous (label-free) contrast provided by second harmonic generation (SHG) microscopy, it is possible to identify early molecular changes occurring in the microtubules of living neurons under ischemic conditions. This is done by measuring the intensity modulation of the SHG signal as a function of the angular rotation of the incident linearly polarized excitation light (technique referred to as PSHG). Our experiments were performed in microtubules from healthy control cultured cortical neurons and were compared to those upon application of several periods of oxygen and glucose deprivation (up to 120 min) causing ischemia. After 120-min oxygen and glucose deprivation, a change in the SHG response to the polarization was measured. Then, by using a three-dimensional PSHG biophysical model, we correlated this finding with the structural changes occurring in the microtubules under oxygen and glucose deprivation. To our knowledge, this is the first study performed in living neuronal cells that is based on direct imaging of axons and that provides the means of identifying the early symptoms of ischemia. Live observation of this process might bring new insights into understanding the dynamics and the mechanisms underlying neuronal degeneration or mechanisms of protection or regeneration.


Subject(s)
Microtubules/ultrastructure , Nerve Degeneration/pathology , Neurons/ultrastructure , Optical Imaging/methods , Animals , Cell Hypoxia , Microscopy, Confocal/methods , Microscopy, Polarization/methods , Rats , Rats, Sprague-Dawley
9.
Nanomedicine ; 8(8): 1263-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22841914

ABSTRACT

Integrin-binding, Arg-Gly-Asp (RGD)-containing peptides are the most widely used agents to deliver drugs, nanoparticles, and imaging agents. Although in nature, several protein-mediated signal transduction events depend on RGD motifs, the potential of RGD-empowered materials in triggering undesired cell-signaling cascades has been neglected. Using an RGD-functionalized protein nanoparticle, we show here that the RGD motif acts as a powerful trophic factor, supporting extracellular signal-regulated kinase 1/2 (ERK1/2)-linked cell proliferation and partial differentiation of PC12 cells, a neuronlike model. FROM THE CLINICAL EDITOR: This work focuses on RGD peptides, which are among the most commonly used tags for targeted drug delivery. They also promote protoneurite formation and expression of neuronal markers (MAP2) in model PC12 cells, which is an unexpected but relevant event in the functionalization of drugs and their nanocarriers.


Subject(s)
Integrins , Nanoparticles/chemistry , Oligopeptides/chemistry , Peptides/chemistry , Binding Sites , Cell Differentiation , Cell Proliferation , Drug Delivery Systems , Humans , Integrins/chemistry , Integrins/metabolism , Ligands , MAP Kinase Signaling System , Neurons/cytology , Neurons/metabolism , Protein Binding , Signal Transduction
10.
Nanomedicine (Lond) ; 6(6): 1047-61, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21651444

ABSTRACT

UNLABELLED: AIM & METHODS: We have produced two chimerical peptides of 10.2 kDa, each contain four biologically active domains, which act as building blocks of protein-based nonviral vehicles for gene therapy. In solution, these peptides tend to aggregate as amorphous clusters of more than 1000 nm, while the presence of DNA promotes their architectonic reorganization as mechanically stable nanometric spherical entities of approximately 80 nm that penetrate mammalian cells through arginine-glycine-aspartic acid cell-binding domains and promote significant transgene expression levels. RESULTS & CONCLUSION: The structural analysis of the protein in these hybrid nanoparticles indicates a molecular conformation with predominance of α-helix and the absence of cross-molecular, ß-sheet-supported protein interactions. The nanoscale organizing forces generated by DNA-protein interactions can then be observed as a potentially tunable, critical factor in the design of protein-only based artificial viruses for gene therapy.


Subject(s)
DNA/chemistry , DNA/metabolism , Peptides/chemistry , Peptides/metabolism , Proteins/chemistry , Proteins/metabolism , Animals , Cells, Cultured , Circular Dichroism , DNA/genetics , DNA/ultrastructure , Genetic Therapy/methods , Mass Spectrometry , Microscopy, Electron, Transmission , Peptides/genetics , Protein Binding , Proteins/genetics , Proteins/ultrastructure , Rats , Rats, Sprague-Dawley
11.
Opt Express ; 17(16): 14418-25, 2009 Aug 03.
Article in English | MEDLINE | ID: mdl-19654849

ABSTRACT

In this paper we provide, for the first time to our knowledge, the effective orientation of the SHG source in cultured cortical neuronal processes in vitro. This is done by the use of the polarization sensitive second harmonic generation (PSHG) imaging microscopy technique. By performing a pixel-level resolution analysis we found that the SHG dipole source has a distribution of angles centered at thetae =33.96 degrees , with a bandwidth of Deltathetae = 12.85 degrees . This orientation can be related with the molecular geometry of the tubulin heterodimmer contained in microtubules.


Subject(s)
Cerebral Cortex/cytology , Image Enhancement/instrumentation , Microscopy, Fluorescence/instrumentation , Neurons/cytology , Optical Devices , Animals , Computer Simulation , Computer-Aided Design , Equipment Design , Equipment Failure Analysis , Image Enhancement/methods , Light , Microscopy, Fluorescence/methods , Models, Theoretical , Rats , Rats, Sprague-Dawley , Scattering, Radiation
12.
J Neurosci Res ; 87(5): 1240-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19006087

ABSTRACT

Human cerebral calcification has been related to deregulation of intracellular calcium homeostasis. In rat basal ganglia, nimodipine and TMB-8, two commonly used calcium antagonists, worsen the chronic AMPA-induced lesion, whereas only nimodipine potentiates calcification. To investigate whether similar effects are present in the hippocampus, AMPA dose-response and calcium movement blockade were performed. A dose-related increase of both hippocampal lesion and calcification was evident in a saturable mode, mostly different from the continuous globus pallidus response previously observed. The value of 2.7 nmol AMPA, selected as yielding 60% of maximum calcification, was coinjected with nimodipine or/and TMB-8 to determine their influence on tissue damage. TMB-8 increased the AMPA lesion in terms of calcified area, and nimodipine reversed this increase, with no effect alone. These results, divergent from those for the globus pallidus, reveal differences in extra- and intracellular calcium movement between the two neurodegenerative processes. Future work focused on other brain areas is required to understand how control of calcium stores may influence neurodegenerative disease evolution.


Subject(s)
Calcium Channel Blockers/pharmacology , Gallic Acid/analogs & derivatives , Hippocampus/drug effects , Neurons/drug effects , Nimodipine/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , Analysis of Variance , Animals , Anthraquinones , Autoradiography , Calcinosis/chemically induced , Dose-Response Relationship, Drug , Gallic Acid/pharmacology , Hippocampus/pathology , Hippocampus/physiopathology , Male , Monoamine Oxidase/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism
13.
Glia ; 57(1): 93-107, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18709661

ABSTRACT

Short interfering RNA (siRNA) inhibits the synthesis of specific proteins through RNA interference (RNAi). However, siRNA can induce innate immune responses that are mediated by toll-like receptors (TLRs) in cells of the immune system. Here, we sought to evaluate whether siRNA can induce such responses in glial cells. We examined the effects of various siRNA sequences prepared with lipids (oligofectamine). Lipid-siRNA induced variable degrees of silencing-independent nonspecific effects, e.g. increased Stat1 and Cox-2 expression and release of IL-6 and IP-10 in primary astroglia. This was prevented through chemical modification of siRNA by nucleoside 2'-O-methylation, without impairing specific gene silencing. Lipid-siRNA also induced nonspecific responses in purified astroglia, but not in microglia, or 3T3 cells. The highest TLR7 and TLR3 mRNA expression was found in microglia and purified astroglia, respectively. Accordingly, the TLR3 agonist poly(I:C) (PIC) induced higher release of IFN-beta in primary and purified astroglia than in microglia. As siRNA, PIC induced IP-10, Stat1, VCAM-1, and Cox-2 and increased TLR3 mRNA expression. The effects of lipid-siRNA in purified astrocytes were attenuated after silencing TLR3 or TLR7 expression, and by the PKR inhibitor 2-aminopurine. Furthermore, lipid-siRNA induced the expression of RIG-I. In contrast, siRNA devoid of lipids did not enter the astrocytes, did not silence gene expression, and did not induce Stat1 or Cox-2. The results show that, in astroglia, lipid-siRNA induces innate immune responses that are mediated, at least in part, by intracellular mechanism dependent on TLR7, TLR3, and helicases.


Subject(s)
Astrocytes/immunology , Immunity, Innate , Lipids/administration & dosage , RNA Interference/immunology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/immunology , Animals , Cells, Cultured , Immunity, Innate/genetics , Lipids/immunology , Mice , NIH 3T3 Cells
14.
Biochem J ; 411(3): 667-77, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18215131

ABSTRACT

Cerebral ischaemia causes long-lasting protein synthesis inhibition that is believed to contribute to brain damage. Energy depletion promotes translation inhibition during ischaemia, and the phosphorylation of eIF (eukaryotic initiation factor) 2alpha is involved in the translation inhibition induced by early ischaemia/reperfusion. However, the molecular mechanisms underlying prolonged translation down-regulation remain elusive. NMDA (N-methyl-D-aspartate) excitotoxicity is also involved in ischaemic damage, as exposure to NMDA impairs translation and promotes the synthesis of NO (nitric oxide), which can also inhibit translation. In the present study, we investigated whether NO was involved in NMDA-induced protein synthesis inhibition in neurons and studied the underlying molecular mechanisms. NMDA and the NO donor DEA/NO (diethylamine-nitric oxide sodium complex) both inhibited protein synthesis and this effect persisted after a 30 min exposure. Treatments with NMDA or NO promoted calpain-dependent eIF4G cleavage and 4E-BP1 (eIF4E-binding protein 1) dephosphorylation and also abolished the formation of eIF4E-eIF4G complexes; however, they did not induce eIF2alpha phosphorylation. Although NOS (NO synthase) inhibitors did not prevent protein synthesis inhibition during 30 min of NMDA exposure, they did abrogate the persistent inhibition of translation observed after NMDA removal. NOS inhibitors also prevented NMDA-induced eIF4G degradation, 4E-BP1 dephosphorylation, decreased eIF4E-eIF4G-binding and cell death. Although the calpain inhibitor calpeptin blocked NMDA-induced eIF4G degradation, it did not prevent 4E-BP1 dephosphorylation, which precludes eIF4E availability, and thus translation inhibition was maintained. The present study suggests that eIF4G integrity and hyperphosphorylated 4E-BP1 are needed to ensure appropriate translation in neurons. In conclusion, our data show that NO mediates NMDA-induced persistent translation inhibition and suggest that deficient eIF4F activity contributes to this process.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4G/metabolism , N-Methylaspartate/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/drug effects , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Cytoprotection/drug effects , Eukaryotic Initiation Factors , Intracellular Signaling Peptides and Proteins , Neurons/drug effects , Nitric Oxide Synthase/metabolism , Phosphorylation/drug effects , Protein Binding , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology
15.
J Neurochem ; 92(3): 505-18, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659221

ABSTRACT

Janus kinases/STAT pathway mediates cellular responses to certain oxidative stress stimuli and cytokines. Here we examine the activation of Stat1 and Stat3 in rat astrocyte cultures and its involvement in cell death. H(2)O(2), interferon (INF)-gamma and interleukin (IL)-6 but not IL-10 caused cell death. Stat1 was phosphorylated on tyrosine (Tyr)-701 after exposure to H(2)O(2), INF-gamma or IL-6 but not IL-10. Tyr-705 pStat3 was observed after H(2)O(2), IL-6 and IL-10. Also, H(2)O(2) induced serine (Ser)-727 phosphorylation of Stat1 but not Stat3. The degree of Tyr-701 pStat1 by the different treatments positively correlated with the corresponding reduction of cell viability. AG490, a Jak2 inhibitor, prevented Tyr-701 but not Ser-727, Stat1 phosphorylation. Also, AG490 inhibited Tyr-705 Stat3 phosphorylation induced by H(2)O(2) and IL-6 but did not prevent that induced by IL-10. Furthermore, AG490 conferred strong protection against cell death induced by INF-gamma, IL-6 and H(2)O(2). These results suggest that Jak2/Stat1 activation mediates cell death induced by proinflammatory cytokines and peroxides. However, we found evidence suggesting that AG490 reduces oxidative stress induced by H(2)O(2), which further shows that H(2)O(2) and/or derived reactive oxygen species directly activate Jak2/Stat1, but masks the actual involvement of this pathway in H(2)O(2)-induced cell death.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Cytokines/toxicity , DNA-Binding Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Tyrphostins/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Cytoprotection/drug effects , Hydrogen Peroxide/toxicity , Inflammation Mediators/toxicity , Interferon-gamma/toxicity , Interleukin-10/toxicity , Interleukin-6/toxicity , Janus Kinase 2 , Oxidants/toxicity , Oxidative Stress/drug effects , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor , STAT3 Transcription Factor , Signal Transduction/drug effects
16.
J Neuropathol Exp Neurol ; 63(4): 338-49, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15099024

ABSTRACT

Matrix metalloproteinase-3 (MMP-3) degrades components of the extracellular matrix and may participate in the pathogenesis of stroke. Here we examine the expression, activation, and cellular location of MMP-3 and the cleavage of agrin, an MMP-3 substrate, following transient middle cerebral artery occlusion in the rat. MMP-3 was activated by ischemia/reperfusion, which was revealed by the appearance of a cleaved form and increased degradation of a substrate. MMP-3 was observed in ischemic neurons, oligodendrocytes, microvasculature, and reactive microglia/macrophages. In cell cultures, MMP-3 expression was observed in neurons and, to a lesser extent, in mature oligodendrocytes, but not in oligodendrocyte progenitors, astrocytes, or microglia. Casein zymography revealed MMP-3 in cultured neurons. Agrin was expressed in cultured neurons and cultured astrocytes. In brain tissue, agrin was detected in neurons, and following ischemia it was also detected in reactive astrocytes. Addition of MMP-3 to protein extracts from control brain caused neuronal agrin degradation. Following ischemia/reperfusion, agrin disappeared from the tissue membrane fraction and a cleaved agrin fragment was found in tissue protein extracts. The present results show MMP-3 activation and neuronal transmembrane agrin cleavage after ischemia/reperfusion. In addition, the finding that MMP-3 cleaves brain agrin strongly suggests that ischemia-induced MMP-3 activation causes agrin cleavage.


Subject(s)
Agrin/metabolism , Brain Ischemia/metabolism , Enzyme Activation/physiology , Matrix Metalloproteinase 3/metabolism , Reperfusion Injury/metabolism , Animals , Astrocytes/metabolism , Blotting, Western , Brain Ischemia/physiopathology , Cells, Cultured , Immunohistochemistry , Male , Microglia/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Rats , Reperfusion Injury/physiopathology
17.
Neurochem Int ; 44(4): 287-91, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14602092

ABSTRACT

Acute injection of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) into the rat globus pallidus leads to calcium precipitation, neuronal death and gliosis. In order to determine whether L-type calcium channels and/or release of Ca(2+) from intracellular stores contribute to the effects of AMPA, nimodipine and 8-(N,N-diethylamino) octyl-3,4,5-trimethoxybenzoate hydrochloride (TMB-8) were administered in combination with AMPA. Nimodipine, but not TMB-8, tended to exacerbate the calcification process initiated by AMPA; the AMPA/nimodipine/TMB-8 combination produced much more calcium deposition than AMPA (+62%, P<0.05). AMPA alone induced a slight but not significant astroglial reaction. Nimodipine slightly enhanced the astroglial reaction triggered by AMPA, whereas TMB-8 doubled it (P<0.001 versus AMPA). These data suggest that blockade of L-type calcium channels by nimodipine enhances calcium imbalance triggered by AMPA, and the calcium release from the endoplasmic reticulum does not participate in the AMPA-induced calcification.


Subject(s)
Basal Ganglia/drug effects , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Nimodipine/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , Animals , Basal Ganglia/pathology , Drug Synergism , Male , Rats , Rats, Sprague-Dawley
18.
J Neurochem ; 85(6): 1455-67, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787065

ABSTRACT

Apolipoprotein E (apoE) and apoJ are lipid carriers produced in the brain primarily by glial cells. A variety of glial-activating stimuli induce a parallel upregulation of both apolipoproteins expression in vivo and in vitro. To further characterize the cell type and mechanisms by which apoE and apoJ expression are upregulated in activated glia, mixed glial cultures from neonatal rat cortex were treated with the endotoxin lipopolysaccharide (LPS). LPS induced dose-dependent increases in apoJ and decreases in apoE expression and secretion with maximum effects at 1-10 ng/mL and 0.1-1 microg/mL, respectively. Experiments with enriched astroglial and microglial cultures demonstrated that apoE and apoJ expression are predominantly microglial and astroglial, respectively. Given the pivotal role that nuclear factor-kappa B (NF-kappa B) plays in glial activation, we assessed its possible role in mediating apoE and apoJ expression by activated glia. LPS robustly increased NF-kappa B activation in mixed glial cultures. Two NF-kappa B inhibitors, aspirin (10 mM) and MG-132 (0.1 microM), blocked basal apoE and apoJ secretion as well as LPS-induced apoJ secretion. These data demonstrate that glial apoE and apoJ expression are independently regulated by LPS in microglia and astroglia, respectively, and that activated microglia are the predominant source of apoE in mixed glial cultures. The transcription factor NF-kappa B appears to be a critical mediator of LPS-stimulated apoJ expression from astroglia.


Subject(s)
Apolipoproteins E/biosynthesis , Astrocytes/metabolism , Glycoproteins/biosynthesis , Lipopolysaccharides/pharmacology , Microglia/metabolism , Molecular Chaperones/biosynthesis , Animals , Animals, Newborn , Apolipoproteins E/genetics , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Clusterin , Coculture Techniques , Cyclooxygenase Inhibitors/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Glycoproteins/genetics , Microglia/cytology , Microglia/drug effects , Molecular Chaperones/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
19.
J Biol Chem ; 278(32): 29552-9, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12771152

ABSTRACT

Transforming growth factor-alpha (TGF-alpha), a ligand of the epidermal growth factor receptor, reduces the infarct size after focal cerebral ischemia in rat, but the molecular basis underlying the protection is unknown. Excitotoxicity and global inhibition of translation are acknowledged to contribute significantly to the ischemic damage. Here we studied whether TGF-alpha can rescue neurons from excitotoxicity in vitro and how it affects calcium homeostasis, protein synthesis, and the associated Akt and extracellular signal-regulated kinase 1/2 (Erk1/2) intracellular signaling pathways in mixed neuron-glia cortical cultures. We found that 100 ng/ml TGF-alpha attenuated neuronal cell death induced by a 30-min exposure to 35 microM N-methyl-D-aspartic acid (NMDA) (as it reduced lactate dehydrogenase release, propidium iodide staining, and caspase-3 activation) and decreased the elevation of intracellular Ca2+ elicited by NMDA. TGF-alpha induced a prompt and sustained phosphorylation of Erk1/2 and prevented the loss of Akt-P induced by NMDA 3 h after exposure. The protective effect of TGF-alpha was completely prevented by PD 98059, an inhibitor of the Erk1/2 pathway. Studies of incorporation of [3H]leucine into proteins showed that NMDA decreased the rate of protein synthesis, and TGF-alpha attenuated this effect. TGF-alpha stimulated the phosphorylation of the eukaryotic initiation factor 4E (eIF4E) but did not affect eIF2 alpha, two proteins involved in translation regulation. PD 98059 abrogated the TGF-alpha effect on eIF4E. Our data demonstrate that TGF-alpha exerts a neuroprotective action against NMDA toxicity, in which Erk1/2 activation plays a key role, and suggest that the underlying mechanisms involve recovery of translation inhibition, mediated at least in part by eIF4E phosphorylation.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , N-Methylaspartate/toxicity , Transforming Growth Factor alpha/physiology , Animals , Blotting, Western , Calcium/metabolism , Caspase 3 , Caspases/metabolism , Cell Death , Cell Nucleus/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Immunohistochemistry , Leucine/chemistry , Mitogen-Activated Protein Kinase 3 , Neurons/drug effects , Neurons/pathology , Phosphorylation , Propidium/pharmacology , Protein Biosynthesis , Rats , Signal Transduction , Time Factors , Transforming Growth Factor alpha/metabolism
20.
J Neurochem ; 83(6): 1338-48, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12472888

ABSTRACT

Induction of heat shock proteins (HSPs) protects cells from oxidative injury. Here Hsp72, Hsp27 and heme oxygenase-1 (HO-1) were induced in cultured rat astrocytes, and protection against oxidative stress was investigated. Astrocytes were treated with sodium arsenite (20-50 micro m) for 1 h, which was non-toxic to cells, 24 h later they were exposed to 400 micro m H2O2 for 1 h, and cell death was evaluated at different time points. Arsenite triggered strong induction of HSPs, which was prevented by 1 micro g/mL cycloheximide (CXH). H2O2 caused cell loss and increased cell death with features of apoptosis, i.e. TdT-mediated dUTP nick-end labelling (TUNEL) reaction and caspase-3 activation. These features were abrogated by pre-treatment with arsenite, which prevented cell loss and significantly reduced the number of dead cells. The protective effect of arsenite was not detected in the presence of CHX. Pre-treatment with arsenite increased protein kinase B (Akt) and extracellular signal regulated kinase 1/2 (ERK1/2) phosphorylation after H2O2. However, while Akt phosphorylation was prevented by CHX, Erk1/2 phosphorylation was further enhanced by CHX. The results show that transient arsenite pre-treatment induces Hsp72, HO-1 and, to a lesser extent, Hsp27; it reduces H2O2-induced astrocyte death; and it causes selective activation of Akt following H2O2. It is suggested that HSP expression at the time of H2O2 exposure protects astrocytes from oxidative injury and apoptotic cell death by means of pro-survival Akt.


Subject(s)
Arsenites/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/toxicity , Protein Serine-Threonine Kinases , Sodium Compounds/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/cytology , Caspase 3 , Caspases/metabolism , Cells, Cultured , Enzyme Activation/drug effects , HSP27 Heat-Shock Proteins , HSP72 Heat-Shock Proteins , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , In Situ Nick-End Labeling , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Oxidants/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphorylation/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...