Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Dec 30.
Article in English | MEDLINE | ID: mdl-38234780

ABSTRACT

Signal-dependent RNA Polymerase II (Pol2) productive elongation is an integral component of gene transcription, including those of immediate early genes (IEGs) induced by neuronal activity. However, it remains unclear how productively elongating Pol2 overcome nucleosomal barriers. Using RNAi, three degraders, and several small molecule inhibitors, we show that the mammalian SWI/SNF complex of neurons (neuronal BAF, or nBAF) is required for activity-induced transcription of neuronal IEGs, including Arc . The nBAF complex facilitates promoter-proximal Pol2 pausing, signal-dependent Pol2 recruitment (loading), and importantly, mediates productive elongation in the gene body via interaction with the elongation complex and elongation-competent Pol2. Mechanistically, Pol2 elongation is mediated by activity-induced nBAF assembly (especially, ARID1A recruitment) and its ATPase activity. Together, our data demonstrate that the nBAF complex regulates several aspects of Pol2 transcription and reveal mechanisms underlying activity-induced Pol2 elongation. These findings may offer insights into human maladies etiologically associated with mutational interdiction of BAF functions.

2.
J Biol Chem ; 298(9): 102278, 2022 09.
Article in English | MEDLINE | ID: mdl-35863435

ABSTRACT

Immediate early genes (IEGs) are transcribed in response to neuronal activity from sensory stimulation during multiple adaptive processes in the brain. The transcriptional profile of IEGs is indicative of the duration of neuronal activity, but its sensitivity to the strength of depolarization remains unknown. Also unknown is whether activity history of graded potential changes influence future neuronal activity. In this work with dissociated rat cortical neurons, we found that mild depolarization-mediated by elevated extracellular potassium (K+)-induces a wide array of rapid IEGs and transiently depresses transcriptional and signaling responses to a successive stimulus. This latter effect was independent of de novo transcription, translation, and signaling via calcineurin or mitogen-activated protein kinase. Furthermore, as measured by multiple electrode arrays and calcium imaging, mild depolarization acutely subdues subsequent spontaneous and bicuculline-evoked activity via calcium- and N-methyl-d-aspartate receptor-dependent mechanisms. Collectively, this work suggests that a recent history of graded potential changes acutely depress neuronal intrinsic properties and subsequent responses. Such effects may have several potential downstream implications, including reducing signal-to-noise ratio during synaptic plasticity processes.


Subject(s)
Action Potentials , Calcineurin , Genes, Immediate-Early , Neurons , Transcription, Genetic , Action Potentials/drug effects , Action Potentials/physiology , Animals , Bicuculline/pharmacology , Calcineurin/genetics , Calcineurin/metabolism , Calcium/metabolism , GABA-A Receptor Antagonists/pharmacology , Genes, Immediate-Early/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/physiology , Potassium/metabolism , Potassium/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
3.
J Biol Chem ; 296: 100024, 2021.
Article in English | MEDLINE | ID: mdl-33410399

ABSTRACT

The human genome contains vast genetic diversity as naturally occurring coding variants, yet the impact of these variants on protein function and physiology is poorly understood. RGS14 is a multifunctional signaling protein that suppresses synaptic plasticity in dendritic spines of hippocampal neurons. RGS14 also is a nucleocytoplasmic shuttling protein, suggesting that balanced nuclear import/export and dendritic spine localization are essential for RGS14 functions. We identified genetic variants L505R (LR) and R507Q (RQ) located within the nuclear export sequence (NES) of human RGS14. Here we report that RGS14 encoding LR or RQ profoundly impacts protein functions in hippocampal neurons. RGS14 membrane localization is regulated by binding Gαi-GDP, whereas RGS14 nuclear export is regulated by Exportin 1 (XPO1). Remarkably, LR and RQ variants disrupt RGS14 binding to Gαi1-GDP and XPO1, nucleocytoplasmic equilibrium, and capacity to inhibit long-term potentiation (LTP). Variant LR accumulates irreversibly in the nucleus, preventing RGS14 binding to Gαi1, localization to dendritic spines, and inhibitory actions on LTP induction, while variant RQ exhibits a mixed phenotype. When introduced into mice by CRISPR/Cas9, RGS14-LR protein expression was detected predominantly in the nuclei of neurons within hippocampus, central amygdala, piriform cortex, and striatum, brain regions associated with learning and synaptic plasticity. Whereas mice completely lacking RGS14 exhibit enhanced spatial learning, mice carrying variant LR exhibit normal spatial learning, suggesting that RGS14 may have distinct functions in the nucleus independent from those in dendrites and spines. These findings show that naturally occurring genetic variants can profoundly alter normal protein function, impacting physiology in unexpected ways.


Subject(s)
Cell Nucleus/metabolism , Hippocampus/metabolism , Long-Term Potentiation , Mutation , Neurons/metabolism , RGS Proteins/genetics , Animals , Hippocampus/cytology , Hippocampus/physiology , Humans , Karyopherins/metabolism , Mice , Neuronal Plasticity , Protein Transport , RGS Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Spatial Learning , Exportin 1 Protein
4.
ASN Neuro ; 12: 1759091420974807, 2020.
Article in English | MEDLINE | ID: mdl-33256465

ABSTRACT

Elevated extracellular potassium chloride is widely used to achieve membrane depolarization of cultured neurons. This technique has illuminated mechanisms of calcium influx through L-type voltage sensitive calcium channels, activity-regulated signaling, downstream transcriptional events, and many other intracellular responses to depolarization. However, there is enormous variability in these treatments, including durations from seconds to days and concentrations from 3mM to 150 mM KCl. Differential effects of these variable protocols on neuronal activity and transcriptional programs are underexplored. Furthermore, potassium chloride treatments in vitro are criticized for being poor representatives of in vivo phenomena and are questioned for their effects on cell viability. In this review, we discuss the intracellular consequences of elevated extracellular potassium chloride treatment in vitro, the variability of such treatments in the literature, the strengths and limitations of this tool, and relevance of these studies to brain functions and dysfunctions.


Subject(s)
Action Potentials/drug effects , Action Potentials/physiology , Neuromuscular Depolarizing Agents/pharmacology , Neurons/drug effects , Neurons/physiology , Potassium Chloride/pharmacology , Animals , Calcium Channels, L-Type/physiology , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology
5.
J Biol Chem ; 295(18): 6120-6137, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32229587

ABSTRACT

The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body ß-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.


Subject(s)
Ethers/chemistry , Ethers/pharmacology , Halogenation , Nervous System/growth & development , Neurons/cytology , Neurons/drug effects , Signal Transduction/drug effects , Animals , Drosophila melanogaster , Hydroxylation , Intracellular Space/drug effects , Intracellular Space/metabolism , Nervous System/cytology , Nervous System/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
6.
Article in English | MEDLINE | ID: mdl-31362383

ABSTRACT

Disruption of epigenetic regulation by environmental toxins is an emerging area of focus for understanding the latter's impact on human health. Polybrominated diphenyl ethers (PBDEs), one such group of toxins, are an environmentally pervasive class of brominated flame retardants that have been extensively used as coatings on a wide range of consumer products. Their environmental stability, propensity for bioaccumulation, and known links to adverse health effects have evoked extensive research to characterize underlying biological mechanisms of toxicity. Of particular concern is the growing body of evidence correlating human exposure levels to behavioral deficits related to neurodevelopmental disorders. The developing nervous system is particularly sensitive to influence by environmental signals, including dysregulation by toxins. Several major modes of actions have been identified, but a clear understanding of how observed effects relate to negative impacts on human health has not been established. Here, we review the current body of evidence for PBDE-induced epigenetic disruptions, including DNA methylation, chromatin dynamics, and non-coding RNA expression while discussing the potential relationship between PBDEs and neurodevelopmental disorders.


Subject(s)
Environmental Pollutants/toxicity , Epigenesis, Genetic/drug effects , Flame Retardants/toxicity , Halogenated Diphenyl Ethers/toxicity , DNA Methylation , Humans
7.
BMC Genomics ; 20(1): 477, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31185909

ABSTRACT

BACKGROUND: Global RNA sequencing technologies have revealed widespread RNA polymerase II (Pol II) transcription outside of gene promoters. Small 5'-capped RNA sequencing (Start-seq) originally developed for the detection of promoter-proximal Pol II pausing has helped improve annotation of Transcription Start Sites (TSSs) of genes as well as identification of non-genic regulatory elements. However, apart from the most well studied genomes of human and mouse, mammalian transcription has not been profiled with sufficiently high precision. RESULTS: We prepared and sequenced Start-seq libraries from rat (Rattus norgevicus) primary neural progenitor cells. Over 48 million uniquely mappable reads from two independent biological replicates allowed us to define the TSSs of 7365 known genes in the rn6 genome, reannotating 2503 TSSs by more than 5 base pairs, characterize promoter-associated antisense transcription, and profile Pol II pausing. By combining TSS data with polyA-selected RNA sequencing, we also identified thousands of potential new genes producing stable RNA as well as non-genic transcripts representing possible regulatory elements. CONCLUSIONS: Our study has produced the first Start-seq dataset for the rat. Apart from profiling transcription initiation, our data reaffirm the prevalence of Pol II pausing across the rat genome and indicate conservation of pausing mechanisms across metazoan genomes. We suggest that pausing location, at least in mammals, is constrained by a distance from initiation of transcription, whether it occurs at or outside of a gene promoter. Abundant antisense transcription initiation around protein coding genes indicates that Pol II recruited to the vicinity of a promoter is distributed to available start sites of transcription at either DNA strand. Transcriptome profiling of neural progenitors presented here will facilitate further studies of other rat cell types as well as other organisms.


Subject(s)
Genomics , Neural Stem Cells/metabolism , RNA Polymerase II/metabolism , Transcription Initiation, Genetic , Animals , Female , Pregnancy , RNA, Antisense/genetics , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Transcription Initiation Site
8.
Environ Epigenet ; 4(1): dvx020, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29765770

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) are a pervasive class of brominated flame retardants that are present in the environment at particularly high levels, especially in the United States. Their environmental stability, propensity for bioaccumulation, and known potential for neurotoxicity has evoked interest regarding their effects on the developing nervous system. Exposure to PBDEs has been strongly associated with neurodevelopmental disorders. However, the details of their mechanistic roles in such disorders are incompletely understood. Here, we report the effects of one of the most prevalent congeners, BDE-47, and its hydroxylated metabolites on the maturation and function of embryonic rat cortical neurons. Prolonged exposure to 6OH-BDE-47 produces the strongest effects amongst the parent BDE-47 congener and its tested hydroxylated metabolites. These effects include: i) disruption of transcriptional responses to neuronal activity, ii) dysregulation of multiple genes associated with neurodevelopmental disorders, and intriguingly, iii) altered expression of several subunits of the developmentally-relevant BAF (Brg1-associated factors) chromatin remodeling complex, including the key subunit BAF170. Taken together, our data indicate that persistent exposure to 6OH-BDE-47 may interfere with neurodevelopmental chromatin remodeling mechanisms and gene transcription programs, which in turn are likely to interfere with downstream processes such as synapse development and overall functional maturity of neurons. Results from this study have identified a novel aspect of 6OH-BDE-47 toxicity and open new avenues to explore the effects of a ubiquitous environmental toxin on epigenetic regulation of neuronal maturation and function.

9.
Neuron ; 98(3): 530-546.e11, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29681534

ABSTRACT

A vast number of different neuronal activity patterns could each induce a different set of activity-regulated genes. Mapping this coupling between activity pattern and gene induction would allow inference of a neuron's activity-pattern history from its gene expression and improve our understanding of activity-pattern-dependent synaptic plasticity. In genome-scale experiments comparing brief and sustained activity patterns, we reveal that activity-duration history can be inferred from gene expression profiles. Brief activity selectively induces a small subset of the activity-regulated gene program that corresponds to the first of three temporal waves of genes induced by sustained activity. Induction of these first-wave genes is mechanistically distinct from that of the later waves because it requires MAPK/ERK signaling but does not require de novo translation. Thus, the same mechanisms that establish the multi-wave temporal structure of gene induction also enable different gene sets to be induced by different activity durations.


Subject(s)
Cerebral Cortex/physiology , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Neurons/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Photic Stimulation/methods , Rats , Rats, Sprague-Dawley
10.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28856239

ABSTRACT

The histone variant H2A.Z is an essential and conserved regulator of eukaryotic gene transcription. However, the exact role of this histone in the transcriptional process remains perplexing. In vertebrates, H2A.Z has two hypervariants, H2A.Z.1 and H2A.Z.2, that have almost identical sequences except for three amino acid residues. Due to such similarity, functional specificity of these hypervariants in neurobiological processes, if any, remain largely unknown. In this study with dissociated rat cortical neurons, we asked if H2A.Z hypervariants have distinct functions in regulating basal and activity-induced gene transcription. Hypervariant-specific RNAi and microarray analyses revealed that H2A.Z.1 and H2A.Z.2 regulate basal expression of largely nonoverlapping gene sets, including genes that code for several synaptic proteins. In response to neuronal activity, rapid transcription of our model gene Arc is impaired by depletion of H2A.Z.2, but not H2A.Z.1. This impairment is partially rescued by codepletion of the H2A.Z chaperone, ANP32E. In contrast, under a different context (after 48 h of tetrodotoxin, TTX), rapid transcription of Arc is impaired by depletion of either hypervariant. Such context-dependent roles of H2A.Z hypervariants, as revealed by our multiplexed gene expression assays, are also evident with several other immediate early genes, where regulatory roles of these hypervariants vary from gene to gene under different conditions. Together, our data suggest that H2A.Z hypervariants have context-specific roles that complement each other to mediate activity-induced neuronal gene transcription.


Subject(s)
Cytoskeletal Proteins/metabolism , Histones/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synaptic Transmission/physiology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Epigenesis, Genetic , Gene Expression Regulation/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/genetics , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Rats, Sprague-Dawley , Transcription, Genetic/physiology
11.
Nat Neurosci ; 14(7): 848-56, 2011 May 29.
Article in English | MEDLINE | ID: mdl-21623364

ABSTRACT

Transcription of immediate early genes (IEGs) in neurons is highly sensitive to neuronal activity, but the mechanism underlying these early transcription events is largely unknown. We found that several IEGs, such as Arc (also known as Arg3.1), are poised for near-instantaneous transcription by the stalling of RNA polymerase II (Pol II) just downstream of the transcription start site in rat neurons. Depletion through RNA interference of negative elongation factor, a mediator of Pol II stalling, reduced the Pol II occupancy of the Arc promoter and compromised the rapid induction of Arc and other IEGs. In contrast, reduction of Pol II stalling did not prevent transcription of IEGs that were expressed later and largely lacked promoter-proximal Pol II stalling. Together, our data strongly indicate that the rapid induction of neuronal IEGs requires poised Pol II and suggest a role for this mechanism in a wide variety of transcription-dependent processes, including learning and memory.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation/physiology , Immediate-Early Proteins/metabolism , Muscle Proteins/metabolism , Neurons/metabolism , RNA Polymerase II/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , Anesthetics, Local/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Cerebral Cortex/cytology , Chromatin Immunoprecipitation/methods , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Exons/drug effects , Exons/physiology , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Immediate-Early Proteins/genetics , Muscle Proteins/genetics , Neurons/drug effects , Oligonucleotide Array Sequence Analysis/methods , Phosphopyruvate Hydratase/metabolism , RNA Interference/physiology , RNA Polymerase II/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Serine/metabolism , Tetrodotoxin/pharmacology , Time Factors , Transcription Factors/metabolism
12.
J Immunol ; 183(3): 2068-78, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19596989

ABSTRACT

Despite being a proinflammatory cytokine, TNF-alpha preconditions neurons against various toxic insults. However, underlying molecular mechanisms are poorly understood. The present study identifies the importance of CREB-binding protein (CBP) in facilitating TNF-alpha-mediated preconditioning in neurons. Treatment of rat primary neurons with fibrillar amyloid beta1-42 (Abeta) resulted in the loss of CBP protein. However, this loss was compensated by TNF-alpha preconditioning as the expression of neuronal CBP was up-regulated in response to TNF-alpha treatment. The induction of CBP by TNF-alpha was observed only in neurons, but not in astroglia and microglia, and it was contingent on the activation of transcription factor NF-kappaB. Interestingly, antisense knockdown of CBP abrogated the TNF-alpha-mediated preconditioning of neurons against Abeta and glutamate toxicity. Similarly in vivo, preadministration of TNF-alpha in mouse neocortex prevented Abeta-induced apoptosis and loss of choline acetyltransferase-positive cholinergic neurons. However, coadministration of cbp antisense, but not scrambled oligonucleotides, negated the protective effect of TNF-alpha against Abeta neurotoxicity. This study illustrates a novel biological role of TNF-alpha in increasing neuron-specific expression of CBP for preconditioning that may have therapeutic potential against neurodegenerative disorders.


Subject(s)
CREB-Binding Protein/genetics , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Cells, Cultured , NF-kappa B/metabolism , Neocortex , Neurons/cytology , Rats , Up-Regulation
13.
Exp Biol Med (Maywood) ; 233(4): 385-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18367626

ABSTRACT

The neuronal nucleus is now widely accepted as playing a vital role in maintaining long-term changes in synaptic effectiveness. To act, however, the nucleus must be appropriately relayed with information regarding the latest round of synaptic plasticity. Several constraints of doing so in a neuron pertain to the often significant spatial distance of synapses from the nucleus and the number of synapses required for such a signal to reach functional levels in the nucleus. Largely based on the sensitivity of transcriptional responses to NMDA receptor antagonists, it has been postulated that the signals are physically relayed by biochemical messengers from the synapse to the nucleus. Alternatively, a second, less often considered but equally viable method of signal transduction may be initiated by action potentials generated proximal to the nucleus, wherefrom the signal can be relayed directly by calcium or indirectly by biochemical second messengers. We consider action potential-dependent signaling to the nucleus to have its own computational advantages over the synapse-to-nucleus signal for some functions. This minireview summarizes the logic and experimental support for these two modes of signaling and attempts to validate the action potential model as playing an important role in transcriptional regulation relating specifically to long-term synaptic plasticity.


Subject(s)
Action Potentials/physiology , Cell Nucleus/metabolism , Neurons , Animals , Calcium/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/physiology , Signal Transduction/physiology , Transcription, Genetic
14.
J Immunol ; 179(10): 7101-9, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982102

ABSTRACT

MAPK-p38 plays an important role in inflammation. Several studies have shown that blocking p38 activity attenuates the transcriptional activity of the proinflammatory transcription factor NF-kappaB without altering its DNA-binding activity. We have also observed that blocking p38 in human primary astrocytes suppresses the transcriptional but not the DNA-binding activity of NF-kappaB and down-regulates the expression of an NF-kappaB-dependent gene, inducible NO synthase. However, the molecular mechanism of p38-mediated regulation of NF-kappaB remains largely unknown. In this study, we delineate that p38 controls the transcriptional activity of NF-kappaB by regulating acetylation of p65, but not its phosphorylation. The combination of IL-1beta and IFN-gamma, previously shown to strongly induce inducible NO synthase in human primary astrocytes, induced p38-dependent phosphorylation of acetyltransferase coactivator p300, but not p65, and subsequent association of p300 with p65. Furthermore, immunocomplex-histone acetyltransferase assays demonstrated that cytokine-induced association of p65 with biologically active immunocomplex-histone acetyltransferase assay was dependent on p38. It has been previously reported that acetylation of p65 at K310 residue is important for transcriptional activity of NF-kappaB. Accordingly, we found that cytokine-induced association of p65 with p300 led to acetylation of p65 at K310. Because p38 regulated the association between p65 and p300, blocking p38 activity also led to attenuation of p65-K310 acetylation in cytokine-stimulated astrocytes. Taken together, this study illuminates a novel regulatory role of p38 during neuroinflammation where this MAP kinase controls acetylation of NF-kappaB p65 by regulating acetyltransferase activity of coactivator p300.


Subject(s)
Astrocytes/immunology , Nitric Oxide Synthase Type II/immunology , Protein Processing, Post-Translational/immunology , Transcription Factor RelA/immunology , p300-CBP Transcription Factors/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Acetylation/drug effects , Antiviral Agents/pharmacology , Astrocytes/metabolism , Astrocytes/pathology , Cells, Cultured , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Nitric Oxide Synthase Type II/biosynthesis , Protein Processing, Post-Translational/drug effects , Transcription Factor RelA/metabolism , Transcription, Genetic/immunology , p300-CBP Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Free Radic Biol Med ; 42(12): 1866-78, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17512466

ABSTRACT

HIV-associated dementia, like several other neurodegenerative diseases, is characterized by selective degeneration of neurons amidst survival of glial cells like astroglia. The molecular basis of such selective susceptibility within the same milieu remains largely unknown. Neurons are rarely infected by the virus. However, they are vulnerable to viral products, like HIV-1 coat protein gp120. Interestingly, gp120 induced oxidative stress in neurons, but not in astroglia. This led us to postulate that astroglia were armed with a more efficient antioxidant system than neurons. Here, we report that the constitutive level of MnSOD (SOD2), the major cellular antioxidant enzyme, is significantly higher in astroglia than in neurons. Furthermore, gp120 treatment enhanced MnSOD levels in astroglia but decreased the same in neurons. This increase in astroglial MnSOD was dependent on NF-kappaB, the crucial transcription factor required for sod2 gene transcription. Blocking NF-kappaB with p65-antisense, p65-si-RNA, or a specific inhibitor, NBD peptide, led to reduced MnSOD levels and enhanced vulnerability of astroglia to gp120. Additionally, neurons were found to have a lower constitutive level of NF-kappaB p65 than astrocytes. Overexpression of p65 increased the level of MnSOD in neurons. This, in turn, elicited greater neuronal resistance to gp120. Taken together, our study suggests that astroglia manifest a higher threshold for gp120-induced lethality than neurons due to greater MnSOD availability, which is demonstrated due to greater level of NF-kappaB p65.


Subject(s)
AIDS Dementia Complex/metabolism , Astrocytes/enzymology , HIV Envelope Protein gp120/pharmacology , NF-kappa B/metabolism , Neurons/enzymology , Superoxide Dismutase/metabolism , AIDS Dementia Complex/pathology , Animals , Antioxidants/pharmacology , Apoptosis/physiology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Female , Fetus/embryology , Fetus/metabolism , Fluorescent Antibody Technique , Humans , Immunoblotting , In Situ Nick-End Labeling , Mitochondria/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neurons/drug effects , Oxidative Stress , Pregnancy , Pregnancy Trimester, Second , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
16.
Neurochem Int ; 49(2): 154-63, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16740341

ABSTRACT

Nitric oxide (NO), being a double-edged sword depending on its concentration in the microenvironment, is involved in both physiological and pathological processes of many organ systems including brain and spinal cord. It is now well-documented that once inducible nitric oxide synthase (iNOS) is expressed in CNS in a signal-dependent fashion, NO in excess of physiological thresholds is produced and this excess NO then plays a role in the pathogenesis of stroke, demyelination and other neurodegenerative diseases. Therefore, a keen interest has been generated in recent years in comprehending the regulation of this enzyme in brain cells. The present review summarizes our current understanding of signaling mechanisms leading to transcription of the iNOS gene in activated astrocytes. We attempt this comprehension with a hope to identify potential targets to intervene NO-mediated CNS disorders.


Subject(s)
Astrocytes/enzymology , Brain Diseases/enzymology , Central Nervous System/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Animals , Brain Diseases/physiopathology , Central Nervous System/physiopathology , Humans , Lipid Metabolism/physiology , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics
17.
Antioxid Redox Signal ; 8(5-6): 929-47, 2006.
Article in English | MEDLINE | ID: mdl-16771683

ABSTRACT

Elevated levels of NO produced within the central nervous system (CNS) are associated with the pathogenesis of neuroinflammatory and neurodegenerative human diseases such as multiple sclerosis, HIV dementia, brain ischemia, trauma, Parkinson's disease, and Alzheimer's disease. Resident glial cells in the CNS (astroglia and microglia) express inducible nitric oxide synthase (iNOS) and produce high levels of NO in response to a wide variety of proinflammatory and degenerative stimuli. Although pathways resulting in the expression of iNOS may vary in two different glial cells of different species, the intracellular signaling events required for the expression of iNOS in these cells are slowly becoming clear. Various signaling cascades converge to activate several transcription factors that control the transcription of iNOS in glial cells. The present review summarizes different results and discusses current understandings about signaling mechanisms for the induction of iNOS expression in activated glial cells. A complete understanding of the regulation of iNOS expression in glial cells is expected to identify novel targets for therapeutic intervention in NO-mediated neurological disorders.


Subject(s)
Gene Expression Regulation, Enzymologic , Neuroglia/enzymology , Nitric Oxide Synthase Type II/metabolism , Animals , Bacteria/metabolism , Cyclic AMP/metabolism , Cytokines/metabolism , Humans , Mevalonic Acid/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type II/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Viruses/metabolism
18.
J Neuroimmune Pharmacol ; 1(3): 212-22, 2006 Sep.
Article in English | MEDLINE | ID: mdl-18040799

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is widely known to be involved in physiological and pathophysiological processes of the brain where this proinflammatory cytokine is implicated with regulation of inflammatory and survival components. We report that TNF-alpha up-regulates exon-IV-bdnf mRNA and brain-derived neurotrophic factor (BDNF) protein in primary astrocytes. The BDNF protein was detectable both in cellular lysate and in the extracellular medium. Activation of NF-kappaB by TNF-alpha and inhibition of TNF-alpha-induced BDNF expression by Deltap65 (a dominant-negative mutant) and NEMO-binding domain peptide (an inhibitor of NF-kappaB) suggests that TNF-alpha induces BDNF expression through the activation of NF-kappaB. Similarly, TNF-alpha induced the activation of C/EBPbeta and the expression of BDNF was sensitive to overexpression of DeltaC/EBPbeta (a dominant-negative mutant) and ETO (an inhibitor of C/EBPbeta). Among three MAP kinases, TNF-alpha-induced BDNF up-regulation was sensitive only to inhibitors of ERK MAP kinase. However, the ERK MAP kinase pathway was coupled to activation of C/EBPbeta but not NF-kappaB. Taken together, this study identifies a novel property of TNF-alpha in inducing the expression of BDNF via NF-kappaB and C/EBPbeta in astrocytes that may be responsible for neurotrophic activity of the cytokine.


Subject(s)
Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neuroimmunomodulation/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Immunohistochemistry , NF-kappa B/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
19.
Free Radic Biol Med ; 38(5): 655-64, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15683721

ABSTRACT

The present study was undertaken to investigate the mechanism of expression of inducible nitric oxide synthase (iNOS) in human primary astrocytes. Among IL-1beta, TNF-alpha, and IFN-gamma, only IL-1beta alone was capable of inducing iNOS. Similarly, among different cytokine combinations, the combinations involving only IL-1beta as a partner were capable of inducing iNOS. The combination of IL-1beta and IFN-gamma (IL-IF) induced the expression of iNOS at the highest level. All three cytokines alone induced the activation of AP-1 while IL-1beta and TNF-alpha but not IFN-gamma induced the activation of NF-kappaB. However, among the three cytokines, only IL-1beta was capable of inducing the activation of CCAAT/enhancer-binding proteinbeta (C/EBPbeta), suggesting an essential role of C/EBPbeta in the expression of iNOS in astrocytes. Although IL-1beta and IFN-gamma alone induced the activation of AP-1, the combination of these two cytokines (IL-IF) markedly inhibited the activation of AP-1. Consistently, JNK-I, a specific inhibitor of JNK, inhibited IL-1beta-mediated activation of AP-1 and expression of iNOS. On the other hand, JNK-I had no effect on (IL-IF)-induced expression of iNOS, suggesting that the activation of AP-1 is involved only during the low level of iNOS induction by IL-1beta but not during the high level of induction by IL-IF. In contrast, the activation of gamma-activation site (GAS) was involved only during the high level of induction by IL-IF but not during the low level of induction by IL-1beta. However, the activation of NF-kappaB and C/EBPbeta was involved in the induction of iNOS by IL-1beta as well as by IL-IF.


Subject(s)
Astrocytes/enzymology , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Cytokines/pharmacology , Nitric Oxide Synthase/biosynthesis , Astrocytes/drug effects , Enzyme Induction , Gene Expression Regulation, Enzymologic , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Interleukin-1beta , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Lipopolysaccharides/pharmacology , NF-kappa B/physiology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II , Peptide Fragments/pharmacology , Promoter Regions, Genetic/physiology , Signal Transduction/physiology , Transcription Factor AP-1/physiology , Tumor Necrosis Factor-alpha/pharmacology
20.
FEBS Lett ; 563(1-3): 223-8, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15063753

ABSTRACT

Environmental factor(s), such as viral infection, has been implicated as one of the triggering events leading to neuroinflammation in multiple sclerosis. This study underlines the importance of double-stranded RNA (dsRNA), the active component of a viral infection, in inducing the expression of inducible nitric oxide synthase (iNOS) in human astroglia. DsRNA in the form of synthetic polyinosinic-polycytidylic acid (poly IC) induced expression of iNOS and iNOS promoter-driven luciferase activity through activation of nuclear factor (NF)-kappaB and CCAAT/enhancer-binding proteinbeta (C/EBPbeta). In addition, we show that inhibitors of protein kinase R attenuated iNOS by suppressing the activation of NF-kappaB but not C/EBPbeta. In contrast, knock down of p38 mitogen-activated protein kinase (MAPK) attenuated iNOS by suppressing the activation of C/EBPbeta but not NF-kappaB. This study delineates a novel role of dsRNA in inducing the expression of iNOS through dsRNA-activated protein kinase (PKR)-mediated activation of NF-kappaB and p38-mediated activation of C/EBPbeta in human astroglia that may participate in virus-induced neurological abnormalities.


Subject(s)
Astrocytes/enzymology , Nitric Oxide Synthase/metabolism , RNA, Double-Stranded/metabolism , eIF-2 Kinase/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Cells, Cultured , Enzyme Activation , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II , Poly I-C/pharmacology , Promoter Regions, Genetic , Time Factors , Transcriptional Activation , Transfection , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...