Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
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.
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
4.
Biochemistry ; 54(21): 3286-301, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25996083

ABSTRACT

Turnover of mRNA is a critical step that allows cells to control gene expression. Endoribonucleases, enzymes cleaving RNA molecules internally, are some of the key components of the degradation process. Here we provide a detailed characterization of novel endoribonuclease SLFN14 purified from rabbit reticulocyte lysate. Schlafen genes encode a family of proteins limited to mammals. Their cellular function is unknown or incompletely understood. In reticulocytes, SLFN14 is strongly overexpressed, represented exclusively by the short form, all tethered to ribosomes, and appears to be one of the major ribosome-associated proteins. SLFN14 binds to ribosomes and ribosomal subunits in the low part of the body and cleaves RNA but preferentially rRNA and ribosome-associated mRNA. This results in the degradation of ribosomal subunits. This process is strictly Mg(2+)- and Mn(2+)-dependent, NTP-independent, and sequence nonspecific. However, in other cell types, SLFN14 is a full-length solely nuclear protein, which lacks ribosomal binding and nuclease activities. Mutational analysis revealed the ribosomal binding site and the aspartate essential for the endonucleolytic activity of protein. Only few endoribonucleases participating in ribosome-mediated processes have been characterized to date. Moreover, none of them are shown to be directly associated with the ribosome. Therefore, our findings expand the general knowledge of endoribonucleases involved in mammalian translation control.


Subject(s)
Endoribonucleases/metabolism , RNA, Ribosomal/metabolism , Rabbits/metabolism , Reticulocytes/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Animals , Binding Sites , Endoribonucleases/chemistry , Endoribonucleases/genetics , Mutation , Rabbits/genetics
5.
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
6.
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
7.
PLoS One ; 7(5): e36475, 2012.
Article in English | MEDLINE | ID: mdl-22615770

ABSTRACT

The pathogenesis of inflammation in the central nervous system (CNS), which contributes to numerous neurodegenerative diseases and results in encephalopathy and neuroinflammation, is poorly understood. Sphingolipid metabolism plays a crucial role in maintaining cellular processes in the CNS, and thus mediates the various pathological consequences of inflammation. For a better understanding of the role of sphingosine kinase activation during neuroinflammation, we developed a bacterial lipopolysaccharide (LPS)-induced brain injury model. The onset of the inflammatory response was observed beginning 4 hours after intracerebral injection of LPS into the lateral ventricles of the brain. A comparison of established neuroinflammatory parameters such as white matter rarefactions, development of cytotoxic edema, astrogliosis, loss of oligodendrocytes, and major cytokines levels in wild type and knockout mice suggested that the neuroinflammatory response in SphK1-/- mice was significantly upregulated. At 6 hours after intracerebroventricular injection of LPS in SphK1-/- mice, the immunoreactivity of the microglia markers and astrocyte marker glial fibrillary acidic protein (GFAP) were significantly increased, while the oligodendrocyte marker O4 was decreased compared to WT mice. Furthermore, western blotting data showed increased levels of GFAP. These results suggest that SphK1 activation is involved in the regulation of LPS induced brain injury. RESEARCH HIGHLIGHTS: • Lipopolysaccharide (LPS) intracerebral injection induces severe neuroinflammation. • Sphingosine kinase 1 deletion worsens the effect of the LPS. • Overexpression of SphK1 might be a potential new treatment approach to neuroinflammation.


Subject(s)
Central Nervous System Diseases/chemically induced , Inflammation/chemically induced , Lipopolysaccharides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Base Sequence , DNA Primers , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
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
9.
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
10.
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
11.
Brain Res Mol Brain Res ; 133(1): 110-8, 2005 Jan 05.
Article in English | MEDLINE | ID: mdl-15661371

ABSTRACT

Local protein synthesis in dendrites is thought to provide a mechanism for long-lasting modifications of synapses in response to physiological activity and behavioral experience. New synthesis of dendritic proteins may be triggered by various paradigms, including induction of epileptiform activity. Prerequisite for such modulated synthesis is a mechanism that limits translation of synaptodendritic mRNAs to times of demand. Recently identified as a translational repressor that is localized to dendrites, small untranslated BC1 RNA has been implicated in the regulation of postsynaptic protein synthesis. Here we show that translational repressor BC1 RNA is itself undergoing modulation as a result of neuronal stimulation. Induction of hippocampal epileptiform activity resulted in a significant decrease of BC1 RNA in the CA3 region over several hours after excitation. The observed decrease was cell-wide, thus indicating reduced expression rather than intracellular redistribution. We suggest that a downregulation of the translational repressor BC1 RNA serves to modulate postsynaptic protein complements in response to the induction of epileptiform activity. Such increased protein synthesis in dendrites may be required for the consolidation of enduring epileptogenic mechanisms.


Subject(s)
Dendrites/metabolism , Gene Expression Regulation/physiology , Kindling, Neurologic/metabolism , RNA, Small Cytoplasmic/metabolism , Analysis of Variance , Animals , Autoradiography/methods , Brain/cytology , Brain/physiology , Electric Stimulation , Electroencephalography/methods , Immunohistochemistry/methods , In Situ Hybridization/methods , Kindling, Neurologic/physiology , Male , RNA, Small Cytoplasmic/genetics , Rats , Rats, Sprague-Dawley
12.
J Biol Chem ; 279(50): 52613-22, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15371429

ABSTRACT

Protein kinase Mzeta (PKMzeta) is an atypical protein kinase C isoform that has been implicated in the protein synthesis-dependent maintenance of long term potentiation and memory storage in the brain. Synapse-associated kinases are uniquely positioned to promote enduring consolidation of structural and functional modifications at the synapse, provided that kinase mRNA is available on site for local input-specific translation. We now report that the mRNA encoding PKMzeta is rapidly transported and specifically localized to synaptodendritic neuronal domains. Transport of PKMzeta mRNA is specified by two cis-acting dendritic targeting elements (Mzeta DTEs). Mzeta DTE1, located at the interface of the 5'-untranslated region and the open reading frame, directs somato-dendritic export of the mRNA. Mzeta DTE2, in contrast, is located in the 3'-untranslated region and is required for delivery of the mRNA to distal dendritic segments. Colocalization with translational repressor BC1 RNA in hippocampal dendrites suggests that PKMzeta mRNA may be subject to translational control in local domains. Dendritic localization of PKMzeta mRNA provides a molecular basis for the functional integration of synaptic signal transduction and translational control pathways.


Subject(s)
Dendrites/metabolism , Memory/physiology , Protein Kinase C/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Base Sequence , Biological Transport, Active , Cells, Cultured , Hippocampus/metabolism , Nucleic Acid Conformation , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/metabolism , Rats , Signal Transduction
13.
Carcinogenesis ; 25(11): 2125-33, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15240511

ABSTRACT

BC200 RNA, a small functional RNA that operates as a translational modulator, has been implicated in the regulation of local synaptodendritic protein synthesis in neurons. Cell type-specific expression of BC200 RNA is tightly controlled such that the RNA is not normally detected in somatic cells other than neurons. However, the neuron-specific control of BC200 expression is deregulated in a number of tumors. We here report that BC200 RNA is expressed at high levels in invasive carcinomas of the breast. In normal breast tissue or in benign tumors such as fibroadenomas, in contrast, we found that the RNA is not detectable at significant levels. The difference in expression levels between invasive carcinomas and normal/benign tissue was statistically highly significant. Receiver Operating Characteristics analysis of sensitivity and specificity confirmed the diagnostic power of BC200 RNA as a molecular marker of invasive breast cancer. In ductal carcinomas in situ, furthermore, significant BC200 expression was associated with high nuclear grade, suggesting that the presence of BC200 RNA in such tumors may be used as a prognostic indicator of tumor progression. The combined results demonstrate the potential of BC200 expression to serve as a molecular tool in the diagnosis and/or prognosis of breast cancer.


Subject(s)
Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , RNA, Neoplasm/genetics , Biomarkers, Tumor , Biopsy , Breast/cytology , Breast Neoplasms/surgery , Female , Humans , Neoplasm Invasiveness , Reference Values
14.
Mol Cell Biol ; 23(18): 6435-41, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12944471

ABSTRACT

Despite the potentially important roles of untranslated RNAs in cellular form or function, genes encoding such RNAs have until now received surprisingly little attention. One such gene encodes BC1 RNA, a small non-mRNA that is delivered to dendritic microdomains in neurons. We have now eliminated the BC1 RNA gene in mice. Three independent founder lines were established from separate embryonic stem cells. The mutant mice appeared to be healthy and showed no anatomical or neurological abnormalities. The gross brain morphology was unaltered in such mice, as were the subcellular distributions of two prototypical dendritic mRNAs (encoding MAP2 and CaMKIIalpha). Due to the relatively recent evolutionary origin of the gene, we expected molecular and behavioral consequences to be subtle. Behavioral analyses, to be reported separately, indicate that the lack of BC1 RNA appears to reduce exploratory activity.


Subject(s)
Brain/physiology , Gene Targeting , Neurons/physiology , RNA, Small Cytoplasmic/genetics , RNA, Untranslated/genetics , Animals , Base Sequence , Brain/anatomy & histology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Dendrites/physiology , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Neurons/metabolism , Phenotype , RNA, Messenger/metabolism , Reference Values
15.
J Neurosci ; 22(23): 10232-41, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12451124

ABSTRACT

In neurons, local protein synthesis in synaptodendritic microdomains has been implicated in the growth and plasticity of synapses. Prerequisites for local translation are the targeted transport of RNAs to distal sites of synthesis in dendrites and translational control mechanisms to limit synthesis to times of demand. Here we identify dendritic BC1 RNA as a specific repressor of translation. Experimental use of internal ribosome entry mechanisms and sucrose density gradient centrifugation showed that BC1-mediated repression targets translation at the level of initiation. Specifically, BC1 RNA inhibited formation of the 48S preinitiation complex, i.e., recruitment of the small ribosomal subunit to the messenger RNA (mRNA). However, 48S complex formation that is independent of the eukaryotic initiation factor 4 (eIF4) family of initiation factors was found to be refractory to inhibition by BC1 RNA, a result that implicates at least one of these factors in the BC1 repression pathway. Biochemical experiments indicated a specific interaction of BC1 RNA with eIF4A, an RNA unwinding factor, and with poly(A)-binding protein. Both proteins were found enriched in synaptodendritic microdomains. Significantly, BC1-mediated repression was shown to be effective not only in cap-dependent translation initiation but also in eIF4-dependent internal initiation. The results suggest a functional role of BC1 RNA as a mediator of translational control in local protein synthesis in nerve cells.


Subject(s)
Dendrites/metabolism , Gene Expression Regulation/physiology , Protein Biosynthesis/physiology , RNA, Small Cytoplasmic/metabolism , Repressor Proteins/metabolism , Animals , Brain Chemistry , Cell-Free System , Cells, Cultured , Electrophoretic Mobility Shift Assay , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation/drug effects , Macromolecular Substances , Neuronal Plasticity/physiology , Neurons/cytology , Peptide Chain Initiation, Translational/physiology , Poly(A)-Binding Proteins/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , RNA, Small Cytoplasmic/pharmacology , Rats , Rats, Sprague-Dawley , Ribosomes/metabolism
16.
J Neurosci ; 22(11): 4293-301, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12040034

ABSTRACT

In neurons, localized RNAs have been identified in dendrites and axons; however, RNA transport in axons remains poorly understood. Here we analyzed axonal RNA transport in goldfish Mauthner neurons in vivo. BC1 RNA, a noncoding RNA polymerase III transcript that is targeted to dendrites in neurons of the rodent nervous system, was used as a probe for axonal RNA transport. Somata of Mauthner neurons were microinjected with various RNAs. Full-length BC1 RNA, but not control RNAs of similar length, was targeted to both axons and dendrites of Mauthner neurons. BC1 RNA was transported in the form of a rapidly advancing wave front that progressed along axons, in a microtubule-dependent manner, at a rate of 2 micrometer/sec. Whereas a BC1 5' segment of 65 nucleotides was transported to axons and dendrites in a way indistinguishable from full-length BC1 RNA, a BC1 3' segment of 60 nucleotides did not enter Mauthner cell processes to any significant extent. In the wake of the wave advancing through the axon, BC1 RNA was found localized to discrete, spatially delimited domains at the axonal surface. Such demarcated cortical concentrations of BC1 RNA could not be observed after disruption of F-actin organization in the axon. It is concluded that the specific delivery of BC1 RNA to spatially defined axonal target sites is a two-step process that requires the sequential participation of microtubules for long-range axial transport and of actin filaments for local radial transfer and focal accumulation in cortical domains.


Subject(s)
Axonal Transport/physiology , Axons/metabolism , Neurons/metabolism , RNA, Small Cytoplasmic/metabolism , 5' Untranslated Regions/physiology , Actins/drug effects , Actins/metabolism , Animals , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dendrites/metabolism , Goldfish , Microinjections , Microtubules/metabolism , Neurons/cytology , Time Factors , Vinblastine/pharmacology
17.
J Cell Sci ; 115(Pt 6): 1243-50, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11884523

ABSTRACT

BC1 RNA, a small non-coding RNA polymerase III transcript, is selectively targeted to dendritic domains of a subset of neurons in the rodent nervous system. It has been implicated in the regulation of local protein synthesis in postsynaptic microdomains. The gene encoding BC1 RNA has been suggested to be a master gene for repetitive ID elements that are found interspersed throughout rodent genomes. A prerequisite for the generation of repetitive elements through retroposition and subsequent transmission in the germline is expression of the master gene RNA in germ cells. To test this hypothesis, we have investigated expression of BC1 RNA in murine male germ cells. We report that BC1 RNA is expressed at substantial levels in a subset of male germ cells. Results from cell fractionation experiments, developmental analysis, and northern and in situ hybridization showed that the RNA was expressed in pre-meiotic spermatogonia, with particularly high amounts in syncytial ensembles of cells that are primed for synchronous spermatogenic differentiation. BC1 RNA continued to be expressed in spermatocytes, but expression levels decreased during further spermatogenic development, and low or negligible amounts of BC1 RNA were identified in round and elongating spermatids. The combined data indicate that BC1 RNA operates in groups of interconnected germ cells, including spermatogonia, where it may function in the mediation of translational control. At the same time, the identification of BC1 RNA in germ cells provides essential support for the hypothesis that repetitive ID elements in rodent genomes arose from the BC1 RNA gene through retroposition.


Subject(s)
RNA, Small Cytoplasmic/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/metabolism , Animals , Brain/physiology , Gene Expression Regulation, Developmental , In Situ Hybridization , Male , Mice , Mice, Mutant Strains , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/metabolism , Seminiferous Tubules/ultrastructure , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Testis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...