Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 127: 390-397, 2019 07.
Article in English | MEDLINE | ID: mdl-30928642

ABSTRACT

Tuberous sclerosis (TSC) is an autosomal dominant disorder caused by heterozygous mutations in the TSC1 or TSC2 gene. TSC is often associated with neurological, cognitive, and behavioral deficits. TSC patients also express co-morbidity with anxiety and mood disorders. The mechanism of pathogenesis in TSC is not entirely clear, but TSC-related neurological symptoms are accompanied by excessive glutamatergic activity and altered synaptic spine structures. To address whether extrasynaptic (e)NMDA-type glutamate receptor (NMDAR) antagonists, as opposed to antagonists that block physiological phasic synaptic activity, can ameliorate the synaptic and behavioral features of this disease, we utilized the Tsc2+/- mouse model of TSC to measure biochemical, electrophysiological, histological, and behavioral parameters in the mice. We found that antagonists that preferentially block tonic activity as found at eNMDARs, particularly the newer drug NitroSynapsin, provide biological and statistically significant improvement in Tsc2+/- phenotypes. Accompanying this improvement was correction of activity in the p38 MAPK-TSC-Rheb-mTORC1-S6K1 pathway. Deficits in hippocampal long-term potentiation (LTP), histological loss of synapses, and behavioral fear conditioning in Tsc2+/- mice were all improved after treatment with NitroSynapsin. Taken together, these results suggest that amelioration of excessive excitation, by limiting aberrant eNMDAR activity, may represent a novel treatment approach for TSC.


Subject(s)
Excitatory Amino Acid Antagonists/therapeutic use , Hippocampus/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Tuberous Sclerosis/drug therapy , Animals , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/metabolism , Mice , Mice, Knockout , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
2.
Invest Ophthalmol Vis Sci ; 58(9): 3741-3749, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28738418

ABSTRACT

Purpose: Photoreceptor degeneration in the retina is a major cause of blindness in humans. Elucidating mechanisms of degenerative and neuroprotective pathways in photoreceptors should afford identification and development of therapeutic strategies. Methods: We used mouse genetic models and improved methods for retinal explant cultures. Retinas were enucleated from Mef2d+/+ and Mef2d-/- mice, stained for MEF2 proteins and outer nuclear layer thickness, and assayed for apoptotic cells. Chromatin immunoprecipitation (ChIP) assays revealed MEF2 binding, and RT-qPCR showed levels of transcription factors. We used AAV2 and electroporation to express genes in retinal explants and electroretinograms to assess photoreceptor functionality. Results: We identify a prosurvival MEF2D-PGC1α pathway that plays a neuroprotective role in photoreceptors. We demonstrate that Mef2d-/- mouse retinas manifest decreased expression of PGC1α and increased photoreceptor cell loss, resulting in the absence of light responses. Molecular repletion of PGC1α protects Mef2d-/- photoreceptors and preserves light responsivity. Conclusions: These results suggest that the MEF2-PGC1α cascade may represent a new therapeutic target for drugs designed to protect photoreceptors from developmental- and age-dependent loss.


Subject(s)
Gene Expression Regulation/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Photoreceptor Cells, Vertebrate/physiology , Retinal Degeneration/prevention & control , Aging , Animals , Apoptosis , Cell Survival/physiology , Dependovirus/genetics , Disease Models, Animal , Electroporation , Electroretinography , Female , Genetic Therapy , In Situ Nick-End Labeling , MEF2 Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Real-Time Polymerase Chain Reaction , Retinal Degeneration/genetics , Retinal Degeneration/pathology
3.
Proc Natl Acad Sci U S A ; 114(20): E4048-E4056, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28461502

ABSTRACT

Gaining mechanistic insight into interaction between causative factors of complex multifactorial diseases involving photoreceptor damage might aid in devising effective therapies. Oxidative stress is one of the potential unifying mechanisms for interplay between genetic and environmental factors that contribute to photoreceptor pathology. Interestingly, the transcription factor myocyte enhancer factor 2d (MEF2D) is known to be important in photoreceptor survival, as knockout of this transcription factor results in loss of photoreceptors in mice. Here, using a mild light-induced retinal degeneration model, we show that the diminished MEF2D transcriptional activity in Mef2d+/- retina is further reduced under photostimulation-induced oxidative stress. Reactive oxygen species cause an aberrant redox modification on MEF2D, consequently inhibiting transcription of its downstream target, nuclear factor (erythroid-derived 2)-like 2 (NRF2). NRF2 is a master regulator of phase II antiinflammatory and antioxidant gene expression. In the Mef2d heterozygous mouse retina, NRF2 is not up-regulated to a normal degree in the face of light-induced oxidative stress, contributing to accelerated photoreceptor cell death. Furthermore, to combat this injury, we found that activation of the endogenous NRF2 pathway using proelectrophilic drugs rescues photoreceptors from photo-induced oxidative stress and may therefore represent a viable treatment for oxidative stress-induced photoreceptor degeneration, which is thought to contribute to some forms of retinitis pigmentosa and age-related macular degeneration.


Subject(s)
NF-E2-Related Factor 2/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/etiology , Abietanes , Animals , Disease Models, Animal , Haploinsufficiency , Light/adverse effects , MEF2 Transcription Factors/genetics , Mice , Oxidative Stress , Reactive Oxygen Species/metabolism
4.
Neurobiol Dis ; 84: 99-108, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25796565

ABSTRACT

Nitric oxide (NO) is a gasotransmitter that impacts fundamental aspects of neuronal function in large measure through S-nitrosylation, a redox reaction that occurs on regulatory cysteine thiol groups. For instance, S-nitrosylation regulates enzymatic activity of target proteins via inhibition of active site cysteine residues or via allosteric regulation of protein structure. During normal brain function, protein S-nitrosylation serves as an important cellular mechanism that modulates a diverse array of physiological processes, including transcriptional activity, synaptic plasticity, and neuronal survival. In contrast, emerging evidence suggests that aging and disease-linked environmental risk factors exacerbate nitrosative stress via excessive production of NO. Consequently, aberrant S-nitrosylation occurs and represents a common pathological feature that contributes to the onset and progression of multiple neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's diseases. In the current review, we highlight recent key findings on aberrant protein S-nitrosylation showing that this reaction triggers protein misfolding, mitochondrial dysfunction, transcriptional dysregulation, synaptic damage, and neuronal injury. Specifically, we discuss the pathological consequences of S-nitrosylated parkin, myocyte enhancer factor 2 (MEF2), dynamin-related protein 1 (Drp1), protein disulfide isomerase (PDI), X-linked inhibitor of apoptosis protein (XIAP), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) under neurodegenerative conditions. We also speculate that intervention to prevent these aberrant S-nitrosylation events may produce novel therapeutic agents to combat neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/metabolism , Protein S/metabolism , Animals , Humans
5.
Genom Data ; 3: 24-27, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25485232

ABSTRACT

[Briefly describe the contents of the Data in Brief article. Tell the reader the repository and reference number for the data in the abstract to.] The myocyte enhancer factor 2 (MEF2) family of transcription factors is highly expressed in the brain, and constitutes a key determinant of neuronal survival, differentiation, and synaptic plasticity. However, genome-wide transcriptional profiling of MEF2-regulated genes has not yet been fully elucidated, particularly at the neural stem cell stage. Here we report the results of microarray analysis comparing mRNAs isolated from human neural progenitor/stem cells (hNPCs) derived from embryonic stem cells expressing a control vector versus progenitors expressing a constitutively-active form of MEF2 (MEF2CA), which increases MEF2 activity. Microarray experiments were performed using the Illumina Human HT-12 V4.0 expression beadchip (GEO#: GSE57184). By comparing vector-control cells to MEF2CA cells, microarray analysis identified 1880 unique genes that were differentially expressed. Among these genes, 1121 genes were upregulated and 759 genes were down-regulated. Our results provide a valuable resource for identifying transcriptional targets of MEF2 in hNPCs.

6.
Biochim Biophys Acta ; 1850(8): 1588-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25527866

ABSTRACT

BACKGROUND: Nitric oxide (NO) is a pleiotropic messenger molecule. The multidimensional actions of NO species are, in part, mediated by their redox nature. Oxidative posttranslational modification of cysteine residues to regulate protein function, termed S-nitrosylation, constitutes a major form of redox-based signaling by NO. SCOPE OF REVIEW: S-Nitrosylation directly modifies a number of cytoplasmic and nuclear proteins in neurons. S-Nitrosylation modulates neuronal development by reaction with specific proteins, including the transcription factor MEF2. This review focuses on the impact of S-nitrosylation on neurogenesis and neuronal development. MAJOR CONCLUSIONS: Functional characterization of S-nitrosylated proteins that regulate neuronal development represents a rapidly emerging field. Recent studies reveal that S-nitrosylation-mediated redox signaling plays an important role in several biological processes essential for neuronal differentiation and maturation. GENERAL SIGNIFICANCE: Investigation of S-nitrosylation in the nervous system has elucidated new molecular and cellular mechanisms for neuronal development. S-Nitrosylated proteins in signaling networks modulate key events in brain development. Dysregulation of this redox-signaling pathway may contribute to neurodevelopmental disabilities such as autism spectrum disorder (ASD). Thus, further elucidation of the involvement of S-nitrosylation in brain development may offer potential therapeutic avenues for neurodevelopmental disorders. This article is part of a Special Issue entitled Redox regulation of differentiation and de-differentiation.


Subject(s)
Neurogenesis , Neurons/metabolism , Nitric Oxide/metabolism , Protein Processing, Post-Translational , Brain/cytology , Brain/growth & development , Brain/metabolism , Cysteine/metabolism , Humans , Models, Neurological , Neurons/cytology , Signal Transduction , Transcription Factors/metabolism
7.
Mol Neurodegener ; 9: 48, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25394486

ABSTRACT

Alzheimer's disease (AD) is a devastating disease characterized by synaptic and neuronal loss in the elderly. Compelling evidence suggests that soluble amyloid-ß peptide (Aß) oligomers induce synaptic loss in AD. Aß-induced synaptic dysfunction is dependent on overstimulation of N-methyl-D-aspartate receptors (NMDARs) resulting in aberrant activation of redox-mediated events as well as elevation of cytoplasmic Ca2+, which in turn triggers downstream pathways involving phospho-tau (p-tau), caspases, Cdk5/dynamin-related protein 1 (Drp1), calcineurin/PP2B, PP2A, Gsk-3ß, Fyn, cofilin, and CaMKII and causes endocytosis of AMPA receptors (AMPARs) as well as NMDARs. Dysfunction in these pathways leads to mitochondrial dysfunction, bioenergetic compromise and consequent synaptic dysfunction and loss, impaired long-term potentiation (LTP), and cognitive decline. Evidence also suggests that Aß may, at least in part, mediate these events by causing an aberrant rise in extrasynaptic glutamate levels by inhibiting glutamate uptake or triggering glutamate release from glial cells. Consequent extrasynaptic NMDAR (eNMDAR) overstimulation then results in synaptic dysfunction via the aforementioned pathways. Consistent with this model of Aß-induced synaptic loss, Aß synaptic toxicity can be partially ameliorated by the NMDAR antagonists (such as memantine and NitroMemantine). PSD-95, an important scaffolding protein that regulates synaptic distribution and activity of both NMDA and AMPA receptors, is also functionally disrupted by Aß. PSD-95 dysregulation is likely an important intermediate step in the pathological cascade of events caused by Aß. In summary, Aß-induced synaptic dysfunction is a complicated process involving multiple pathways, components and biological events, and their underlying mechanisms, albeit as yet incompletely understood, may offer hope for new therapeutic avenues.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Synapses/metabolism , Synapses/pathology , Alzheimer Disease/pathology , Animals , Humans
8.
Cell Rep ; 8(1): 217-28, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25001280

ABSTRACT

Redox-mediated posttranslational modifications represent a molecular switch that controls major mechanisms of cell function. Nitric oxide (NO) can mediate redox reactions via S-nitrosylation, representing transfer of an NO group to a critical protein thiol. NO is known to modulate neurogenesis and neuronal survival in various brain regions in disparate neurodegenerative conditions. However, a unifying molecular mechanism linking these phenomena remains unknown. Here, we report that S-nitrosylation of myocyte enhancer factor 2 (MEF2) transcription factors acts as a redox switch to inhibit both neurogenesis and neuronal survival. Structure-based analysis reveals that MEF2 dimerization creates a pocket, facilitating S-nitrosylation at an evolutionally conserved cysteine residue in the DNA binding domain. S-Nitrosylation disrupts MEF2-DNA binding and transcriptional activity, leading to impaired neurogenesis and survival in vitro and in vivo. Our data define a molecular switch whereby redox-mediated posttranslational modification controls both neurogenesis and neurodegeneration via a single transcriptional signaling cascade.


Subject(s)
Apoptosis , MEF2 Transcription Factors/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Nitric Oxide/metabolism , Protein Processing, Post-Translational , Transcriptional Activation , Animals , Binding Sites , Cells, Cultured , DNA/metabolism , HEK293 Cells , Humans , MEF2 Transcription Factors/chemistry , MEF2 Transcription Factors/genetics , Mice , Neural Stem Cells/cytology , Oxidation-Reduction , Protein Binding
9.
J Neurosci ; 34(13): 4640-53, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24672010

ABSTRACT

Mutations in the ataxia telangiectasia mutated (ATM) gene, which encodes a kinase critical for the normal DNA damage response, cause the neurodegenerative disorder ataxia-telangiectasia (AT). The substrates of ATM in the brain are poorly understood. Here we demonstrate that ATM phosphorylates and activates the transcription factor myocyte enhancer factor 2D (MEF2D), which plays a critical role in promoting survival of cerebellar granule cells. ATM associates with MEF2D after DNA damage and phosphorylates the transcription factor at four ATM consensus sites. Knockdown of endogenous MEF2D with a short-hairpin RNA (shRNA) increases sensitivity to etoposide-induced DNA damage and neuronal cell death. Interestingly, substitution of endogenous MEF2D with an shRNA-resistant phosphomimetic MEF2D mutant protects cerebellar granule cells from cell death after DNA damage, whereas an shRNA-resistant nonphosphorylatable MEF2D mutant does not. In vivo, cerebella in Mef2d knock-out mice manifest increased susceptibility to DNA damage. Together, our results show that MEF2D is a substrate for phosphorylation by ATM, thus promoting survival in response to DNA damage. Moreover, dysregulation of the ATM-MEF2D pathway may contribute to neurodegeneration in AT.


Subject(s)
DNA Damage/physiology , Neurons/physiology , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/physiology , Cell Survival/physiology , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Enzyme Inhibitors/pharmacology , Female , HEK293 Cells , Humans , In Vitro Techniques , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , RNA Interference/physiology , Superoxides/metabolism
10.
Cell ; 155(6): 1351-64, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24290359

ABSTRACT

Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.


Subject(s)
Gene-Environment Interaction , Mitochondria/drug effects , Paraquat/toxicity , Parkinson Disease/genetics , Parkinson Disease/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , MEF2 Transcription Factors , Mutation/drug effects , Neurons/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Nitrogen Species/metabolism , Substantia Nigra/metabolism , Transcription Factors/metabolism , Transcription, Genetic , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
11.
Proc Natl Acad Sci U S A ; 110(27): E2518-27, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776240

ABSTRACT

Synaptic loss is the cardinal feature linking neuropathology to cognitive decline in Alzheimer's disease (AD). However, the mechanism of synaptic damage remains incompletely understood. Here, using FRET-based glutamate sensor imaging, we show that amyloid-ß peptide (Aß) engages α7 nicotinic acetylcholine receptors to induce release of astrocytic glutamate, which in turn activates extrasynaptic NMDA receptors (eNMDARs) on neurons. In hippocampal autapses, this eNMDAR activity is followed by reduction in evoked and miniature excitatory postsynaptic currents (mEPSCs). Decreased mEPSC frequency may reflect early synaptic injury because of concurrent eNMDAR-mediated NO production, tau phosphorylation, and caspase-3 activation, each of which is implicated in spine loss. In hippocampal slices, oligomeric Aß induces eNMDAR-mediated synaptic depression. In AD-transgenic mice compared with wild type, whole-cell recordings revealed excessive tonic eNMDAR activity accompanied by eNMDAR-sensitive loss of mEPSCs. Importantly, the improved NMDAR antagonist NitroMemantine, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from Aß-induced damage both in vitro and in vivo.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/metabolism , Glutamic Acid/metabolism , Neural Inhibition/physiology , Peptide Fragments/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Coculture Techniques , Female , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Rats , Receptors, Nicotinic/metabolism , Synapses/metabolism , alpha7 Nicotinic Acetylcholine Receptor
12.
Neuron ; 78(4): 596-614, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23719160

ABSTRACT

S-Nitrosylation is a redox-mediated posttranslational modification that regulates protein function via covalent reaction of nitric oxide (NO)-related species with a cysteine thiol group on the target protein. Under physiological conditions, S-nitrosylation can be an important modulator of signal transduction pathways, akin to phosphorylation. However, with aging or environmental toxins that generate excessive NO, aberrant S-nitrosylation reactions can occur and affect protein misfolding, mitochondrial fragmentation, synaptic function, apoptosis or autophagy. Here, we discuss how aberrantly S-nitrosylated proteins (SNO-proteins) play a crucial role in the pathogenesis of neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. Insight into the pathophysiological role of aberrant S-nitrosylation pathways will enhance our understanding of molecular mechanisms leading to neurodegenerative diseases and point to potential therapeutic interventions.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Reactive Nitrogen Species/metabolism , Animals , Brain/pathology , Humans , Neurodegenerative Diseases/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism
13.
Exp Neurol ; 236(2): 298-306, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22575597

ABSTRACT

HIV/gp120 transgenic mice manifest neuropathological features similar to HIV-associated neurocognitive disorders (HAND) in humans, including astrogliosis, microglia activation, and decreased neuronal synapses. Here, proteomic screening of synaptosomes from HIV/gp120 transgenic mice was conducted to determine potential neuronal markers and drug targets associated with HAND. Synaptosomes from 13 month-old wild-type (wt) and HIV/gp120 transgenic mouse cortex were subjected to tandem mass tag (TMT) labeling and subsequent analysis using an LTQ-Orbitrap mass spectrometer in pulsed-Q dissociation (PQD) mode for tandem mass spectrometry (MS/MS). A total of 1301 proteins were identified in both wt and HIV/gp120 transgenic mice. Three of the most differentially-regulated proteins were validated by immunoblotting. To elucidate putative pathways associated with the proteomic profile, 107 proteins manifesting a ≥1.5 fold change in expression were analyzed using a bioinformatics pathway analysis tool. This analysis revealed direct or indirect involvement of the phosphotidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway, a well-known neuronal survival pathway. Immunoblots confirmed a lower phospho (p)Akt/Akt ratio in synaptosomes from HIV/gp120 transgenic animals compared to wt, suggesting that this neuroprotective pathway was inactivated in the HIV/gp120 transgenic brain. Based on this information, we then compared immunoblots of pAkt/Akt in the forebrains of these mice as well as in human postmortem brain. We observed a significant decrease in the pAkt/Akt ratio in synaptosomes and forebrain of HIV/gp120 transgenic compared to wt mice, and a similar decrease in human forebrain from HAND patients compared to neurologically unimpaired HIV+ and HIV- controls. Moreover, mechanistic insight into an additional pathway for decreased Akt activity in HIV/gp120 mouse brains and human HAND brains was shown to occur via S-nitrosylation of Akt protein, a posttranslational modification known to inhibit Akt activity and contribute to neuronal cell injury and death. Thus, MS proteomic profiling in the HIV/gp120 transgenic mouse predicted dysregulation of the PI3K/Akt pathway observed in human brains with HAND, providing evidence that this mouse is a useful disease model and that the Akt pathway may provide multiple drug targets for the treatment of HIV-related dementias.


Subject(s)
HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp120/genetics , HIV Infections/metabolism , Neurodegenerative Diseases/metabolism , Synaptosomes/physiology , Tandem Mass Spectrometry/methods , Viral Regulatory and Accessory Proteins/genetics , Adult , Aged , Animals , Female , HIV Envelope Protein gp120/physiology , HIV Infections/genetics , HIV Infections/pathology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neural Pathways/chemistry , Neural Pathways/pathology , Neural Pathways/virology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/virology , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/chemistry , Synaptosomes/chemistry , Synaptosomes/virology , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/chemistry , Virus Inactivation
14.
Mol Cell ; 39(2): 184-95, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20670888

ABSTRACT

X-linked inhibitor of apoptosis (XIAP) is a potent antagonist of caspase apoptotic activity. XIAP also functions as an E3 ubiquitin ligase, targeting caspases for degradation. However, molecular pathways controlling XIAP activities remain unclear. Here, we report that nitric oxide (NO) reacts with XIAP by S-nitrosylating its RING domain (forming SNO-XIAP), thereby inhibiting E3 ligase and antiapoptotic activity. NO-mediated neurotoxicity and caspase activation have been linked to several neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's diseases. We find significant SNO-XIAP formation in brains of patients with these diseases, implicating this reaction in the etiology of neuronal damage. Conversely, S-nitrosylation of caspases is known to inhibit apoptotic activity. Unexpectedly, we find that SNO-caspase transnitrosylates (transfers its NO group) to XIAP, forming SNO-XIAP, and thus promotes cell injury and death. These findings provide insights into the regulation of caspase activation in neurodegenerative disorders mediated, at least in part, by nitrosative stress.


Subject(s)
Apoptosis , Caspases/metabolism , Nitric Oxide/metabolism , Protein Processing, Post-Translational , X-Linked Inhibitor of Apoptosis Protein/metabolism , Caspases/genetics , Enzyme Activation/genetics , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Nitric Oxide/genetics , Protein Structure, Tertiary , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics
15.
Nat Med ; 15(12): 1407-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19915593

ABSTRACT

Huntington's disease is caused by an expanded CAG repeat in the gene encoding huntingtin (HTT), resulting in loss of striatal and cortical neurons. Given that the gene product is widely expressed, it remains unclear why neurons are selectively targeted. Here we show the relationship between synaptic and extrasynaptic activity, inclusion formation of mutant huntingtin protein (mtHtt) and neuronal survival. Synaptic N-methyl-D-aspartate-type glutamate receptor (NMDAR) activity induces mtHtt inclusions via a T complex-1 (TCP-1) ring complex (TRiC)-dependent mechanism, rendering neurons more resistant to mtHtt-mediated cell death. In contrast, stimulation of extrasynaptic NMDARs increases the vulnerability of mtHtt-containing neurons to cell death by impairing the neuroprotective cyclic AMP response element-binding protein (CREB)-peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) cascade and increasing the level of the small guanine nucleotide-binding protein Rhes, which is known to sumoylate and disaggregate mtHtt. Treatment of transgenic mice expressing a yeast artificial chromosome containing 128 CAG repeats (YAC128) with low-dose memantine blocks extrasynaptic (but not synaptic) NMDARs and ameliorates neuropathological and behavioral manifestations. By contrast, high-dose memantine, which blocks both extrasynaptic and synaptic NMDAR activity, decreases neuronal inclusions and worsens these outcomes. Our findings offer a rational therapeutic approach for protecting susceptible neurons in Huntington's disease.


Subject(s)
Mutation , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Animals , Cell Death/physiology , Chromosomes, Artificial, Yeast , Huntingtin Protein , Memantine/pharmacology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Nuclear Proteins/genetics , Patch-Clamp Techniques , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/physiology , Transcription Factors
17.
Proc Natl Acad Sci U S A ; 105(27): 9397-402, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18599437

ABSTRACT

Emerging evidence suggests that myocyte enhancer factor 2 (MEF2) transcription factors act as effectors of neurogenesis in the brain, with MEF2C the predominant isoform in developing cerebrocortex. Here, we show that conditional knockout of Mef2c in nestin-expressing neural stem/progenitor cells (NSCs) impaired neuronal differentiation in vivo, resulting in aberrant compaction and smaller somal size. NSC proliferation and survival were not affected. Conditional null mice surviving to adulthood manifested more immature electrophysiological network properties and severe behavioral deficits reminiscent of Rett syndrome, an autism-related disorder. Our data support a crucial role for MEF2C in programming early neuronal differentiation and proper distribution within the layers of the neocortex.


Subject(s)
Cell Differentiation , Myogenic Regulatory Factors/metabolism , Neurons/cytology , Stem Cells/cytology , Transcription Factors/metabolism , Animals , Animals, Newborn , Behavior , Cognition , Electrophysiology , Embryonic Development , MEF2 Transcription Factors , Mice , Mice, Knockout , Mitosis , Neocortex/embryology , Neocortex/pathology , Neurons/pathology , Phenotype
18.
J Neurosci ; 28(26): 6557-68, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18579729

ABSTRACT

Cell-based therapies require a reliable source of cells that can be easily grown, undergo directed differentiation, and remain viable after transplantation. Here, we generated stably transformed murine ES (embryonic stem) cells that express a constitutively active form of myocyte enhancer factor 2C (MEF2CA). MEF2C has been implicated as a calcium-dependent transcription factor that enhances survival and affects synapse formation of neurons as well as differentiation of cardiomyocytes. We now report that expression of MEF2CA, both in vitro and in vivo, under regulation of the nestin enhancer effectively produces "neuronal" progenitor cells that differentiate into a virtually pure population of neurons. Histological, electrophysiological, and behavioral analyses demonstrate that MEF2C-directed neuronal progenitor cells transplanted into a mouse model of cerebral ischemia can successfully differentiate into functioning neurons and ameliorate stroke-induced behavioral deficits.


Subject(s)
Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Myogenic Regulatory Factors/genetics , Stem Cell Transplantation/methods , Stem Cells/metabolism , Transcription Factors/genetics , Animals , Apoptosis/genetics , Brain Ischemia/therapy , Brain Tissue Transplantation/methods , Cell Differentiation/physiology , Cell Line, Transformed , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression Regulation/genetics , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Nerve Degeneration/therapy , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Neurons/metabolism , Organ Culture Techniques , Stem Cells/cytology
19.
J Neurochem ; 104(4): 1116-31, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17995931

ABSTRACT

Electrophilic compounds are a newly recognized class of redox-active neuroprotective compounds with electron deficient, electrophilic carbon centers that react with specific cysteine residues on targeted proteins via thiol (S-)alkylation. Although plants produce a variety of physiologically active electrophilic compounds, the detailed mechanism of action of these compounds remains unknown. Catechol ring-containing compounds have attracted attention because they become electrophilic quinones upon oxidation, although they are not themselves electrophilic. In this study, we focused on the neuroprotective effects of one such compound, carnosic acid (CA), found in the herb rosemary obtained from Rosmarinus officinalis. We found that CA activates the Keap1/Nrf2 transcriptional pathway by binding to specific Keap1 cysteine residues, thus protecting neurons from oxidative stress and excitotoxicity. In cerebrocortical cultures, CA-biotin accumulates in non-neuronal cells at low concentrations and in neurons at higher concentrations. We present evidence that both the neuronal and non-neuronal distribution of CA may contribute to its neuroprotective effect. Furthermore, CA translocates into the brain, increases the level of reduced glutathione in vivo, and protects the brain against middle cerebral artery ischemia/reperfusion, suggesting that CA may represent a new type of neuroprotective electrophilic compound.


Subject(s)
Abietanes/pharmacology , Catechols/pharmacology , Cysteine/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Plant Extracts/pharmacology , Proteins/metabolism , Abietanes/chemistry , Alkylation/drug effects , Animals , COS Cells , Catechols/chemistry , Cattle , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins , Kelch-Like ECH-Associated Protein 1 , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , PC12 Cells , Plant Extracts/chemistry , Rats , Rats, Sprague-Dawley , Rosmarinus , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Cell Stem Cell ; 1(2): 230-6, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-18371353

ABSTRACT

Impaired adult neurogenesis has been observed in several neurodegenerative diseases, including human immunodeficiency virus (HIV-1)-associated dementia (HAD). Here we report that the HIV-envelope glycoprotein gp120, which is associated with HAD pathogenesis, inhibits proliferation of adult neural progenitor cells (aNPCs) in vitro and in vivo in the dentate gyrus of the hippocampus of HIV/gp120-transgenic mice. We demonstrate that HIV/gp120 arrests cell-cycle progression of aNPCs at the G1 phase via a cascade consisting of p38 mitogen-activated protein kinase (MAPK) --> MAPK-activated protein kinase 2 (a cell-cycle checkpoint kinase) --> Cdc25B/C. Our findings define a molecular mechanism that compromises adult neurogenesis in this neurodegenerative disorder.


Subject(s)
HIV Envelope Protein gp120/physiology , HIV Infections/pathology , HIV-1/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Protein Serine-Threonine Kinases/metabolism , Stem Cells/physiology , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/virology , Animals , Cell Growth Processes/physiology , Female , G1 Phase/genetics , G1 Phase/physiology , HIV Envelope Protein gp120/genetics , HIV Infections/enzymology , HIV Infections/virology , MAP Kinase Signaling System , Male , Mice , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/virology , Neurons/cytology , Neurons/enzymology , Rats , cdc25 Phosphatases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...