Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 5(12)2022 10 13.
Article in English | MEDLINE | ID: mdl-36229064

ABSTRACT

Lupus autoimmunity frequently presents with neuropsychiatric manifestations, but underlying etiology remains poorly understood. Human brain cytoplasmic 200 RNA (BC200 RNA) is a translational regulator in neuronal synapto-dendritic domains. Here, we show that a BC200 guanosine-adenosine dendritic transport motif is recognized by autoantibodies from a subset of neuropsychiatric lupus patients. These autoantibodies impact BC200 functionality by quasi irreversibly displacing two RNA transport factors from the guanosine-adenosine transport motif. Such anti-BC autoantibodies, which can gain access to brains of neuropsychiatric lupus patients, give rise to clinical manifestations including seizures. To establish causality, naive mice with a permeabilized blood-brain barrier were injected with anti-BC autoantibodies from lupus patients with seizures. Animals so injected developed seizure susceptibility with high mortality. Seizure activity was entirely precluded when animals were injected with lupus anti-BC autoantibodies together with BC200 decoy autoantigen. Seizures are a common clinical manifestation in neuropsychiatric lupus, and our work identifies anti-BC autoantibody activity as a mechanistic cause. The results demonstrate potential utility of BC200 decoys for autoantibody-specific therapeutic interventions in neuropsychiatric lupus.


Subject(s)
Lupus Vasculitis, Central Nervous System , Adenosine , Animals , Autoantibodies , Autoantigens , Guanosine , Humans , Lupus Vasculitis, Central Nervous System/psychology , Mice , RNA , Seizures
2.
J Neurosci ; 39(39): 7759-7777, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31405929

ABSTRACT

The etiology of the autoimmune disorder systemic lupus erythematosus (SLE) remains poorly understood. In neuropsychiatric SLE (NPSLE), autoimmune responses against neural self-antigens find expression in neurological and cognitive alterations. SLE autoantibodies often target nucleic acids, including RNAs and specifically RNA domains with higher-order structural content. We report that autoantibodies directed against neuronal regulatory brain cytoplasmic (BC) RNAs were generated in a subset of SLE patients. By contrast, anti-BC RNA autoantibodies (anti-BC abs) were not detected in sera from patients with autoimmune diseases other than SLE (e.g., rheumatoid arthritis or multiple sclerosis) or in sera from healthy subjects with no evidence of disease. SLE anti-BC abs belong to the IgG class of immunoglobulins and target both primate BC200 RNA and rodent BC1 RNA. They are specifically directed at architectural motifs in BC RNA 5' stem-loop domains that serve as dendritic targeting elements (DTEs). SLE anti-BC abs effectively compete with RNA transport factor heterogeneous nuclear ribonucleoprotein A2 (hnRNP A2) for DTE access and significantly diminish BC RNA delivery to synapto-dendritic sites of function. In vivo experiments with male BALB/c mice indicate that, upon lipopolysaccharide-induced opening of the blood-brain barrier, SLE anti-BC abs are taken up by CNS neurons where they significantly impede localization of endogenous BC1 RNA to synapto-dendritic domains. Lack of BC1 RNA causes phenotypic abnormalities including epileptogenic responses and cognitive dysfunction. The combined data indicate a role for anti-BC RNA autoimmunity in SLE and its neuropsychiatric manifestations.SIGNIFICANCE STATEMENT Although clinical manifestations of neuropsychiatric lupus are well recognized, the underlying molecular-cellular alterations have been difficult to determine. We report that sera of a subset of lupus patients contain autoantibodies directed at regulatory brain cytoplasmic (BC) RNAs. These antibodies, which we call anti-BC abs, target the BC RNA 5' domain noncanonical motif structures that specify dendritic delivery. Lupus anti-BC abs effectively compete with RNA transport factor heterogeneous nuclear ribonucleoprotein A2 (hnRNP A2) for access to BC RNAs. As a result, hnRNP A2 is displaced, and BC RNAs are impaired in their ability to reach synapto-dendritic sites of function. The results reveal an unexpected link between BC RNA autoantibody recognition and dendritic RNA targeting. Cellular RNA dysregulation may thus be a contributing factor in the pathogenesis of neuropsychiatric lupus.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Lupus Erythematosus, Systemic/immunology , Neurons/metabolism , RNA, Small Cytoplasmic/immunology , RNA, Small Cytoplasmic/metabolism , Animals , Brain/immunology , Brain/metabolism , Female , Humans , Lupus Erythematosus, Systemic/metabolism , Male , Mice , Mice, Inbred BALB C , RNA Transport/physiology
3.
J Investig Med ; 66(7): 1055-1063, 2018 10.
Article in English | MEDLINE | ID: mdl-29967012

ABSTRACT

Regulatory brain cytoplasmic 200 RNA (BC200 RNA) is highly expressed in human mammary carcinoma cells. Here, we ask whether BC200 RNA becomes detectable in peripheral blood of patients with invasive breast cancer. Using quantitative reverse-transcription PCR (qRT-PCR) methodology, we observed that BC200 RNA blood levels were significantly elevated, in comparison with healthy subjects, in patients with invasive breast cancer prior to tumorectomy (p=0.001) and in patients with metastatic breast cancer (p=0.003). In patients with invasive breast cancer who had recently undergone tumorectomy, BC200 RNA blood levels were not distinguishable from levels in healthy subjects. However, normality analysis revealed a heterogeneous distribution of patients in this group, including a subgroup of individuals with high residual BC200 RNA blood levels. In blood from patients with invasive breast cancer, BC200 RNA was specifically detected in the mononuclear leukocyte fraction. The qRT-PCR approach is sensitive enough to detect as few as three BC200 RNA-expressing tumor cells. Our work establishes the potential of BC200 RNA detection in blood to serve as a molecular indicator of invasive breast malignancy.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/genetics , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Middle Aged , Neoplasm Invasiveness , ROC Curve , Sensitivity and Specificity
4.
eNeuro ; 5(2)2018.
Article in English | MEDLINE | ID: mdl-29766042

ABSTRACT

Fragile X premutation disorder is caused by CGG triplet repeat expansions in the 5' untranslated region of FMR1 mRNA. The question of how expanded CGG repeats cause disease is a subject of continuing debate. Our work indicates that CGG-repeat structures compete with regulatory BC1 RNA for access to RNA transport factor hnRNP A2. As a result, BC1 RNA is mislocalized in vivo, as its synapto-dendritic presence is severely diminished in brains of CGG-repeat knock-in animals (a premutation mouse model). Lack of BC1 RNA is known to cause seizure activity and cognitive dysfunction. Our working hypothesis thus predicted that absence, or significantly reduced presence, of BC1 RNA in synapto-dendritic domains of premutation animal neurons would engender cognate phenotypic alterations. Testing this prediction, we established epileptogenic susceptibility and cognitive impairments as major phenotypic abnormalities of CGG premutation mice. In CA3 hippocampal neurons of such animals, synaptic release of glutamate elicits neuronal hyperexcitability in the form of group I metabotropic glutamate receptor-dependent prolonged epileptiform discharges. CGG-repeat knock-in animals are susceptible to sound-induced seizures and are cognitively impaired as revealed in the Attentional Set Shift Task. These phenotypic disturbances occur in young-adult premutation animals, indicating that a neurodevelopmental deficit is an early-initial manifestation of the disorder. The data are consistent with the notion that RNA mislocalization can contribute to pathogenesis.


Subject(s)
Cognitive Dysfunction/genetics , Fragile X Syndrome/genetics , RNA Transport/genetics , RNA, Small Cytoplasmic/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Seizures/genetics , Trinucleotide Repeat Expansion/genetics , Age Factors , Animals , CA3 Region, Hippocampal/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Fragile X Syndrome/complications , Fragile X Syndrome/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/physiology , Seizures/etiology , Seizures/physiopathology
5.
Learn Mem ; 24(7): 267-277, 2017 07.
Article in English | MEDLINE | ID: mdl-28620074

ABSTRACT

Dendritic regulatory BC1 RNA is a non-protein-coding (npc) RNA that operates in the translational control of gene expression. The absence of BC1 RNA in BC1 knockout (KO) animals causes translational dysregulation that entails neuronal phenotypic alterations including prolonged epileptiform discharges, audiogenic seizure activity in vivo, and excessive cortical oscillations in the γ frequency band. Here we asked whether BC1 RNA control is also required for higher brain functions such as learning, memory, or cognition. To address this question, we used odor/object attentional set shifting tasks in which prefrontal cortical performance was assessed in a series of discrimination and conflict learning sessions. Results obtained in these behavioral trials indicate that BC1 KO animals were significantly impaired in their cognitive flexibility. When faced with conflicting information sources, BC1 KO animals committed regressive errors as they were compromised in their ability to disengage from recently acquired memories even though recall of such memories was in conflict with new situational context. The observed cognitive deficits are reminiscent of those previously described in subtypes of human autism spectrum disorders.


Subject(s)
Attention/physiology , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Odorants , RNA, Small Cytoplasmic/metabolism , Animals , Conflict, Psychological , Discrimination Learning/physiology , Grooming/physiology , Learning Curve , Maze Learning , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Cytoplasmic/genetics
6.
J Cell Biol ; 207(2): 237-52, 2014 Oct 27.
Article in English | MEDLINE | ID: mdl-25332164

ABSTRACT

In neurons, translational regulation of gene expression has been implicated in the activity-dependent management of synapto-dendritic protein repertoires. However, the fundamentals of stimulus-modulated translational control in neurons remain poorly understood. Here we describe a mechanism in which regulatory brain cytoplasmic (BC) RNAs cooperate with eukaryotic initiation factor 4B (eIF4B) to control translation in a manner that is responsive to neuronal activity. eIF4B is required for the translation of mRNAs with structured 5' untranslated regions (UTRs), exemplified here by neuronal protein kinase Mζ (PKMζ) mRNA. Upon neuronal stimulation, synapto-dendritic eIF4B is dephosphorylated at serine 406 in a rapid process that is mediated by protein phosphatase 2A. Such dephosphorylation causes a significant decrease in the binding affinity between eIF4B and BC RNA translational repressors, enabling the factor to engage the 40S small ribosomal subunit for translation initiation. BC RNA translational control, mediated via eIF4B phosphorylation status, couples neuronal activity to translational output, and thus provides a mechanistic basis for long-term plastic changes in nerve cells.


Subject(s)
Eukaryotic Initiation Factors/physiology , Neurons/metabolism , RNA, Messenger/metabolism , RNA, Small Cytoplasmic/physiology , 5' Untranslated Regions , Animals , Cell Line , Eukaryotic Initiation Factors/metabolism , Female , Gene Expression Regulation , Male , Mice , Models, Genetic , Neurons/cytology , Neurons/ultrastructure , Phosphorylation , Protein Biosynthesis , RNA, Small Cytoplasmic/metabolism , Rats, Sprague-Dawley , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosome Subunits, Small, Eukaryotic/physiology , Sf9 Cells , Signal Transduction
7.
J Cell Biol ; 205(4): 493-510, 2014 May 26.
Article in English | MEDLINE | ID: mdl-24841565

ABSTRACT

A key determinant of neuronal functionality and plasticity is the targeted delivery of select ribonucleic acids (RNAs) to synaptodendritic sites of protein synthesis. In this paper, we ask how dendritic RNA transport can be regulated in a manner that is informed by the cell's activity status. We describe a molecular mechanism in which inducible interactions of noncanonical RNA motif structures with targeting factor heterogeneous nuclear ribonucleoprotein (hnRNP) A2 form the basis for activity-dependent dendritic RNA targeting. High-affinity interactions between hnRNP A2 and conditional GA-type RNA targeting motifs are critically dependent on elevated Ca(2+) levels in a narrow concentration range. Dendritic transport of messenger RNAs that carry such GA motifs is inducible by influx of Ca(2+) through voltage-dependent calcium channels upon ß-adrenergic receptor activation. The combined data establish a functional correspondence between Ca(2+)-dependent RNA-protein interactions and activity-inducible RNA transport in dendrites. They also indicate a role of genomic retroposition in the phylogenetic development of RNA targeting competence.


Subject(s)
Aminopeptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Neuronal Plasticity/genetics , Neurons/physiology , RNA Transport/physiology , Serine Proteases/genetics , Aminopeptidases/metabolism , Animals , Base Sequence , Biological Transport/physiology , Calcium Channels/genetics , Calcium Signaling/genetics , Dendrites/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Female , Ganglia, Sympathetic/cytology , Genomics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Male , Molecular Sequence Data , Neurons/ultrastructure , Nucleic Acid Conformation , Phylogeny , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Retroelements/genetics , Serine Proteases/metabolism , Tripeptidyl-Peptidase 1 , Tubulin/genetics
8.
Trends Biochem Sci ; 38(1): 47-55, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23218750

ABSTRACT

Translational control of gene expression is instrumental in the regulation of eukaryotic cellular form and function. Neurons in particular rely on this form of control because their numerous synaptic connections need to be independently modulated in an input-specific manner. Brain cytoplasmic (BC) RNAs implement translational control at neuronal synapses. BC RNAs regulate protein synthesis by interacting with eIF4 translation initiation factors. Recent evidence suggests that such regulation is required to control synaptic strength, and that dysregulation of local protein synthesis precipitates neuronal hyperexcitability and a propensity for epileptogenic responses. A similar phenotype results from lack of fragile X mental retardation protein (FMRP), indicating that BC RNAs and FMRP use overlapping and convergent modes of action in neuronal translational regulation.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Gene Expression Regulation , Neurons/metabolism , Protein Biosynthesis , RNA/metabolism , Synapses/metabolism , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Humans , RNA/genetics , Synapses/genetics
9.
Mol Cell Biol ; 31(22): 4538-49, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21930783

ABSTRACT

In higher eukaryotes, increasing evidence suggests, gene expression is to a large degree controlled by RNA. Regulatory RNAs have been implicated in the management of neuronal function and plasticity in mammalian brains. However, much of the molecular-mechanistic framework that enables neuronal regulatory RNAs to control gene expression remains poorly understood. Here, we establish molecular mechanisms that underlie the regulatory capacity of neuronal BC RNAs in the translational control of gene expression. We report that regulatory BC RNAs employ a two-pronged approach in translational control. One of two distinct repression mechanisms is mediated by C-loop motifs in BC RNA 3' stem-loop domains. These C-loops bind to eIF4B and prevent the factor's interaction with 18S rRNA of the small ribosomal subunit. In the second mechanism, the central A-rich domains of BC RNAs target eIF4A, specifically inhibiting its RNA helicase activity. Thus, BC RNAs repress translation initiation in a bimodal mechanistic approach. As BC RNA functionality has evolved independently in rodent and primate lineages, our data suggest that BC RNA translational control was necessitated and implemented during mammalian phylogenetic development of complex neural systems.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factors/metabolism , Protein Biosynthesis , RNA, Small Cytoplasmic/metabolism , RNA/metabolism , Animals , Base Sequence , Gene Expression Regulation , Inverted Repeat Sequences , Mice , Neurons/metabolism , Nucleic Acid Conformation , RNA/genetics , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , RNA, Small Cytoplasmic/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Ribosome Subunits, Small/metabolism , Sequence Analysis, RNA
10.
J Cell Biol ; 194(3): 441-57, 2011 Aug 08.
Article in English | MEDLINE | ID: mdl-21807882

ABSTRACT

In neurons, regulation of gene expression occurs in part through translational control at the synapse. A fundamental requirement for such local control is the targeted delivery of select neuronal mRNAs and regulatory RNAs to distal dendritic sites. The nature of spatial RNA destination codes, and the mechanism by which they are interpreted for dendritic delivery, remain poorly understood. We find here that in a key dendritic RNA transport pathway (exemplified by BC1 RNA, a dendritic regulatory RNA, and protein kinase M ζ [PKMζ] mRNA, a dendritic mRNA), noncanonical purine•purine nucleotide interactions are functional determinants of RNA targeting motifs. These motifs are specifically recognized by heterogeneous nuclear ribonucleoprotein A2 (hnRNP A2), a trans-acting factor required for dendritic delivery. Binding to hnRNP A2 and ensuing dendritic delivery are effectively competed by RNAs with CGG triplet repeat expansions. CGG repeats, when expanded in the 5' untranslated region of fragile X mental retardation 1 (FMR1) mRNA, cause fragile X-associated tremor/ataxia syndrome. The data suggest that cellular dysregulation observed in the presence of CGG repeat RNA may result from molecular competition in neuronal RNA transport pathways.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , 5' Untranslated Regions , Animals , DNA Repeat Expansion , Dendrites/genetics , Dendrites/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Gene Expression Regulation , Nucleic Acid Conformation , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Small Cytoplasmic , Rats , Rats, Sprague-Dawley
11.
PLoS One ; 5(11): e15509, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21124905

ABSTRACT

BACKGROUND: BC RNAs and the fragile X mental retardation protein (FMRP) are translational repressors that have been implicated in the control of local protein synthesis at the synapse. Work with BC1 and Fmr1 animal models has revealed that phenotypical consequences resulting from the absence of either BC1 RNA or FMRP are remarkably similar. To establish functional interactions between BC1 RNA and FMRP is important for our understanding of how local protein synthesis regulates neuronal excitability. METHODOLOGY/PRINCIPAL FINDINGS: We generated BC1-/- Fmr1-/- double knockout (dKO) mice. We examined such animals, lacking both BC1 RNA and FMRP, in comparison with single knockout (sKO) animals lacking either one repressor. Analysis of neural phenotypical output revealed that at least three attributes of brain functionality are subject to control by both BC1 RNA and FMRP: neuronal network excitability, epileptogenesis, and place learning. The severity of CA3 pyramidal cell hyperexcitability was significantly higher in BC1-/- Fmr1-/- dKO preparations than in the respective sKO preparations, as was seizure susceptibility of BC1-/- Fmr1-/- dKO animals in response to auditory stimulation. In place learning, BC1-/- Fmr1-/- dKO animals were severely impaired, in contrast to BC1-/- or Fmr1-/- sKO animals which exhibited only mild deficits. CONCLUSIONS/SIGNIFICANCE: Our data indicate that BC1 RNA and FMRP operate in sequential-independent fashion. They suggest that the molecular interplay between two translational repressors directly impacts brain functionality.


Subject(s)
Brain/physiology , Fragile X Mental Retardation Protein/metabolism , Nerve Net/physiology , RNA, Small Cytoplasmic/metabolism , Animals , Avoidance Learning/physiology , Brain/metabolism , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiology , Electrophysiology/methods , Female , Fragile X Mental Retardation Protein/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/metabolism , RNA, Small Cytoplasmic/genetics
12.
Brain Res ; 1338: 36-47, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20307503

ABSTRACT

Regulatory RNAs are being increasingly investigated in neurons, and important roles in brain function have been revealed. Regulatory RNAs are non-protein-coding RNAs (npcRNAs) that comprise a heterogeneous group of molecules, varying in size and mechanism of action. Regulatory RNAs often exert post-transcriptional control of gene expression, resulting in gene silencing or gene expression stimulation. Here, we review evidence that regulatory RNAs are implicated in neuronal development, differentiation, and plasticity. We will also discuss npcRNA dysregulation that may be involved in pathological states of the brain such as neurodevelopmental disorders, neurodegeneration, and epilepsy.


Subject(s)
Brain Diseases/metabolism , Brain/metabolism , RNA, Untranslated/metabolism , Animals , Brain Diseases/genetics , Gene Expression Regulation , Humans
13.
J Neurosci ; 29(32): 9977-86, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19675232

ABSTRACT

Regulatory RNAs have been suggested to contribute to the control of gene expression in eukaryotes. Brain cytoplasmic (BC) RNAs are regulatory RNAs that control translation initiation. We now report that neuronal BC1 RNA plays an instrumental role in the protein-synthesis-dependent implementation of neuronal excitation-repression equilibria. BC1 repression counter-regulates translational stimulation resulting from synaptic activation of group I metabotropic glutamate receptors (mGluRs). Absence of BC1 RNA precipitates plasticity dysregulation in the form of neuronal hyperexcitability, elicited by group I mGluR-stimulated translation and signaled through the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathway. Dysregulation of group I mGluR function in the absence of BC1 RNA gives rise to abnormal brain function. Cortical EEG recordings from freely moving BC1(-/-) animals show that group I mGluR-mediated oscillations in the gamma frequency range are significantly elevated. When subjected to sensory stimulation, these animals display an acute group I mGluR-dependent propensity for convulsive seizures. Inadequate RNA control in neurons is thus causally linked to heightened group I mGluR-stimulated translation, neuronal hyperexcitability, heightened gamma band oscillations, and epileptogenesis. These data highlight the significance of small RNA control in neuronal plasticity.


Subject(s)
Brain/physiology , Neurons/physiology , RNA, Small Cytoplasmic/metabolism , Receptors, Metabotropic Glutamate/metabolism , Action Potentials , Animals , Disks Large Homolog 4 Protein , Electroencephalography , Fragile X Mental Retardation Protein/biosynthesis , Guanylate Kinases , Hippocampus/physiology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Periodicity , RNA, Small Cytoplasmic/genetics , Seizures/physiopathology , Synapses/physiology
15.
Mol Cell Biol ; 28(9): 3008-19, 2008 May.
Article in English | MEDLINE | ID: mdl-18316401

ABSTRACT

Translational repressors, increasing evidence suggests, participate in the regulation of protein synthesis at the synapse, thus providing a basis for the long-term plastic modulation of synaptic strength. Dendritic BC1 RNA is a non-protein-coding RNA that represses translation at the level of initiation. However, the molecular mechanism of BC1 repression has remained unknown. Here we identify the catalytic activity of eukaryotic initiation factor 4A (eIF4A), an ATP-dependent RNA helicase, as a target of BC1-mediated translational control. BC1 RNA specifically blocks the RNA duplex unwinding activity of eIF4A but, at the same time, stimulates its ATPase activity. BC200 RNA, the primate-specific BC1 counterpart, targets eIF4A activity in identical fashion, as a result decoupling ATP hydrolysis from RNA duplex unwinding. In vivo, BC1 RNA represses translation of a reporter mRNA with 5' secondary structure. The eIF4A mechanism places BC RNAs in a central position to modulate protein synthesis in neurons.


Subject(s)
Dendrites/metabolism , Eukaryotic Initiation Factor-4A/biosynthesis , RNA, Small Cytoplasmic/physiology , 5' Flanking Region , Cell Line , Eukaryotic Initiation Factor-4A/genetics , Humans , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Untranslated/physiology
16.
Proc Natl Acad Sci U S A ; 105(2): 734-9, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18184799

ABSTRACT

The fragile X mental retardation protein (FMRP), the functional absence of which causes fragile X syndrome, is an RNA-binding protein that has been implicated in the regulation of local protein synthesis at the synapse. The mechanism of FMRP's interaction with its target mRNAs, however, has remained controversial. In one model, it has been proposed that BC1 RNA, a small non-protein-coding RNA that localizes to synaptodendritic domains, operates as a requisite adaptor by specifically binding to both FMRP and, via direct base-pairing, to FMRP target mRNAs. Other models posit that FMRP interacts with its target mRNAs directly, i.e., in a BC1-independent manner. Here five laboratories independently set out to test the BC1-FMRP model. We report that specific BC1-FMRP interactions could be documented neither in vitro nor in vivo. Interactions between BC1 RNA and FMRP target mRNAs were determined to be of a nonspecific nature. Significantly, the association of FMRP with bona fide target mRNAs was independent of the presence of BC1 RNA in vivo. The combined experimental evidence is discordant with a proposed scenario in which BC1 RNA acts as a bridge between FMRP and its target mRNAs and rather supports a model in which BC1 RNA and FMRP are translational repressors that operate independently.


Subject(s)
Fragile X Mental Retardation Protein/genetics , RNA, Small Cytoplasmic , Animals , Biotinylation , Brain/metabolism , Gene Expression Regulation , Immunoprecipitation , Mice , Mice, Knockout , Nucleic Acid Hybridization , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry
17.
Proc Natl Acad Sci U S A ; 104(25): 10679-84, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17553964

ABSTRACT

Small untranslated BC1 and BC200 RNAs are translational regulators that are selectively targeted to somatodendritic domains of neurons. They are thought to operate as modulators of local protein synthesis in postsynaptic dendritic microdomains, in a capacity in which they would contribute to the maintenance of long-term synaptic plasticity. Because plasticity failure has been proposed to be a starting point for the neurodegenerative changes that are seen in Alzheimer's disease (AD), we asked whether somatodendritic levels of human BC200 RNA are deregulated in AD brains. We found that in normal aging, BC200 levels in cortical areas were reduced by >60% between the ages of 49 and 86. In contrast, BC200 RNA was significantly up-regulated in AD brains, in comparison with age-matched normal brains. This up-regulation in AD was specific to brain areas that are involved in the disease. Relative BC200 levels in those areas increased in parallel with the progression of AD, as reflected by Clinical Dementia Rating scores. In more advanced stages of the disease, BC200 RNA often assumed a clustered perikaryal localization, indicating that dendritic loss is accompanied by somatic overexpression. Mislocalization and overexpression of BC200 RNA may be reactive-compensatory to, or causative of, synaptodendritic deterioration in AD neurons.


Subject(s)
Aging , Alzheimer Disease/genetics , Dendrites/chemistry , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Aged , Aged, 80 and over , Calibration , Case-Control Studies , Cerebral Cortex/metabolism , Gene Expression Regulation , Humans , In Situ Hybridization , Middle Aged , RNA Probes , RNA, Ribosomal/metabolism
18.
RNA Biol ; 3(4): 133-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17172877

ABSTRACT

Three-dimensional architectural motifs are increasingly recognized as determinants of RNA functionality. We submit that such motifs can encode spatial information. RNAs are targeted to subcellular localities in many eukaryotic cell types, and especially in neuronal and glial cells, RNAs can be transported over long distances to their final destination sites. Such RNAs contain cis-acting long-range targeting elements, and recent evidence suggests that kink-turn motifs within such elements may act as spatial codes to direct transport. Kink-turns are complex RNA motifs that feature double- and single-stranded components and introduce a signature three-dimensional structure into helical stems. We propose that the overall architectural design as well as the individual character--as specified by nucleotide identity and arrangement--of kink-turn motifs can serve as RNA targeting determinants.


Subject(s)
Nucleic Acid Conformation , Open Reading Frames/genetics , RNA/chemistry , RNA/genetics , Animals , Base Sequence , Humans , Molecular Sequence Data
19.
J Cell Biol ; 175(3): 427-39, 2006 Nov 06.
Article in English | MEDLINE | ID: mdl-17074884

ABSTRACT

BC1 RNA is a dendritic untranslated RNA that has been implicated in local translational control mechanisms in neurons. Prerequisite for a functional role of the RNA in synaptodendritic domains is its targeted delivery along the dendritic extent. We report here that the targeting-competent 5' BC1 domain carries two dendritic targeting codes. One code, specifying somatic export, is located in the medial-basal region of the 5' BC1 stem-loop structure. It is defined by an export-determinant stem-bulge motif. The second code, specifying long-range dendritic delivery, is located in the apical part of the 5' stem-loop domain. This element features a GA kink-turn (KT) motif that is indispensable for distal targeting. It specifically interacts with heterogeneous nuclear ribonucleoprotein A2, a trans-acting targeting factor that has previously been implicated in the transport of MBP mRNA in oligodendrocytes and neurons. Our work suggests that a BC1 KT motif encodes distal targeting via the A2 pathway and that architectural RNA elements, such as KT motifs, may function as spatial codes in neural cells.


Subject(s)
5' Untranslated Regions/genetics , Dendrites/metabolism , RNA Transport , RNA, Small Cytoplasmic/genetics , 5' Untranslated Regions/metabolism , Animals , Base Sequence , Cells, Cultured , Drosophila Proteins , Embryo, Mammalian , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Homeodomain Proteins/genetics , Microinjections , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Protein Binding , RNA , RNA, Small Cytoplasmic/metabolism , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion , Trans-Activators/genetics
20.
J Mol Biol ; 356(5): 1118-23, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16412460

ABSTRACT

RNA localization is an important means of post-transcriptional regulation of gene expression in many eukaryotic cell types. In neurons, select RNAs are delivered to postsynaptic dendritic microdomains, a mechanism that is considered a key underpinning in the administration of long-term synaptic plasticity. BC1 RNA is a small untranslated RNA that interacts with translation initiation factors and functions as a translational repressor by targeting assembly of 48S initiation complexes. BC1 RNA is specifically and rapidly transported to dendrites where it is found concentrated in postsynaptic microdomains. The cytoskeletal infrastructure underlying dendritic localization of BC1 RNA has not been investigated. We now report that the dendritic delivery of BC1 RNA is dependent on intact microtubules. In two neuronal cell types, hippocampal neurons and sympathetic neurons in primary culture, disruption of microtubules abolished dendritic localization of BC1 RNA. In contrast, disruption of actin filaments had no significant effect on the somatodendritic distribution of BC1 RNA. It is concluded that the long-range dendritic delivery of BC1 RNA is supported by microtubules. At the same time, a role for actin filaments, while unlikely for long-range BC1 delivery, is not ruled out for short-range local translocation and anchoring at dendritic destination sites.


Subject(s)
Dendrites/metabolism , Microtubules/metabolism , Neurons/metabolism , RNA, Small Cytoplasmic/metabolism , Animals , Antineoplastic Agents/pharmacology , Biological Transport , Cells, Cultured , Colchicine/pharmacology , Cytochalasin D/pharmacology , Dendrites/ultrastructure , Hippocampus/cytology , Neurons/cytology , Neurons/drug effects , Nocodazole/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Rats , Sympathetic Nervous System/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...