Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Cell ; 171(5): 994-1000, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149615

ABSTRACT

Eukaryotic translation is tightly regulated to ensure that protein production occurs at the right time and place. Recent studies on abnormal repeat proteins, especially in age-dependent neurodegenerative diseases caused by nucleotide repeat expansion, have highlighted or identified two forms of unconventional translation initiation: usage of AUG-like sites (near cognates) or repeat-associated non-AUG (RAN) translation. We discuss how repeat proteins may differ due to not just unconventional initiation, but also ribosomal frameshifting and/or imperfect repeat DNA replication, expansion, and repair, and we highlight how research on translation of repeats may uncover insights into the biology of translation and its contribution to disease.


Subject(s)
Neurodegenerative Diseases/genetics , Protein Biosynthesis , Animals , Codon, Initiator , Frameshifting, Ribosomal , Humans , Neurodegenerative Diseases/metabolism , Open Reading Frames , Regulatory Sequences, Ribonucleic Acid , Trinucleotide Repeat Expansion
2.
Cell ; 163(2): 292-300, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26451481

ABSTRACT

Among the three phases of mRNA translation-initiation, elongation, and termination-initiation has traditionally been considered to be rate limiting and thus the focus of regulation. Emerging evidence, however, demonstrates that control of ribosome translocation (polypeptide elongation) can also be regulatory and indeed exerts a profound influence on development, neurologic disease, and cell stress. The correspondence of mRNA codon usage and the relative abundance of their cognate tRNAs is equally important for mediating the rate of polypeptide elongation. Here, we discuss recent results showing that ribosome pausing is a widely used mechanism for controlling translation and, as a result, biological transitions in health and disease.


Subject(s)
Gene Expression Regulation , Peptide Chain Elongation, Translational , Polyribosomes/metabolism , Animals , Codon , Disease/genetics , Humans , Peptide Chain Initiation, Translational , RNA, Messenger/metabolism , RNA, Transfer/metabolism
3.
Annu Rev Cell Dev Biol ; 30: 393-415, 2014.
Article in English | MEDLINE | ID: mdl-25068488

ABSTRACT

The cytoplasmic polyadenylation element binding (CPEB) proteins are sequence-specific mRNA binding proteins that control translation in development, health, and disease. CPEB1, the founding member of this family, has become an important model for illustrating general principles of translational control by cytoplasmic polyadenylation in gametogenesis, cancer etiology, synaptic plasticity, learning, and memory. Although the biological functions of the other members of this protein family in vertebrates are just beginning to emerge, it is already evident that they, too, mediate important processes, such as cancer etiology and higher cognitive function. In Drosophila, the CPEB proteins Orb and Orb2 play key roles in oogenesis and in neuronal function, as do related proteins in Caenorhabditis elegans and Aplysia. We review the biochemical features of the CPEB proteins, discuss their activities in several biological systems, and illustrate how understanding CPEB activity in model organisms has an important impact on neurological disease.


Subject(s)
Protein Biosynthesis , RNA, Messenger/metabolism , mRNA Cleavage and Polyadenylation Factors/physiology , Amyloid/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cellular Senescence , Cytoplasm/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Nervous System/metabolism , Nervous System Diseases/metabolism , Polyadenylation
4.
Nat Rev Neurosci ; 22(4): 209-222, 2021 04.
Article in English | MEDLINE | ID: mdl-33608673

ABSTRACT

Fragile X mental retardation protein (FMRP) is the product of the fragile X mental retardation 1 gene (FMR1), a gene that - when epigenetically inactivated by a triplet nucleotide repeat expansion - causes the neurodevelopmental disorder fragile X syndrome (FXS). FMRP is a widely expressed RNA-binding protein with activity that is essential for proper synaptic plasticity and architecture, aspects of neural function that are known to go awry in FXS. Although the neurophysiology of FXS has been described in remarkable detail, research focusing on the molecular biology of FMRP has only scratched the surface. For more than two decades, FMRP has been well established as a translational repressor; however, recent whole transcriptome and translatome analyses in mouse and human models of FXS have shown that FMRP is involved in the regulation of nearly all aspects of gene expression. The emerging mechanistic details of the mechanisms by which FMRP regulates gene expression may offer ways to design new therapies for FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Neurons/metabolism , Trinucleotide Repeat Expansion , Animals , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Humans , Mice , Neuronal Plasticity/physiology
5.
PLoS Biol ; 21(12): e3002417, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38048343

ABSTRACT

Fragile X syndrome (FXS) is a neurodevelopmental disorder that is often modeled in Fmr1 knockout mice where the RNA-binding protein FMRP is absent. Here, we show that in Fmr1-deficient mice, RNA mis-splicing occurs in several brain regions and peripheral tissues. To assess molecular mechanisms of splicing mis-regulation, we employed N2A cells depleted of Fmr1. In the absence of FMRP, RNA-specific exon skipping events are linked to the splicing factors hnRNPF, PTBP1, and MBNL1. FMRP regulates the translation of Mbnl1 mRNA as well as Mbnl1 RNA auto-splicing. Elevated Mbnl1 auto-splicing in FMRP-deficient cells results in the loss of a nuclear localization signal (NLS)-containing exon. This in turn alters the nucleus-to-cytoplasm ratio of MBNL1. This redistribution of MBNL1 isoforms in Fmr1-deficient cells could result in downstream splicing changes in other RNAs. Indeed, further investigation revealed that splicing disruptions resulting from Fmr1 depletion could be rescued by overexpression of nuclear MBNL1. Altered Mbnl1 auto-splicing also occurs in human FXS postmortem brain. These data suggest that FMRP-controlled translation and RNA processing may cascade into a general dys-regulation of splicing in Fmr1-deficient cells.


Subject(s)
Fragile X Mental Retardation Protein , RNA Splicing , Animals , Humans , Mice , Cytoplasm/metabolism , Fragile X Mental Retardation Protein/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Protein Isoforms/metabolism , RNA/metabolism , RNA Splicing/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
6.
Cell ; 146(2): 247-61, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21784246

ABSTRACT

FMRP loss of function causes Fragile X syndrome (FXS) and autistic features. FMRP is a polyribosome-associated neuronal RNA-binding protein, suggesting that it plays a key role in regulating neuronal translation, but there has been little consensus regarding either its RNA targets or mechanism of action. Here, we use high-throughput sequencing of RNAs isolated by crosslinking immunoprecipitation (HITS-CLIP) to identify FMRP interactions with mouse brain polyribosomal mRNAs. FMRP interacts with the coding region of transcripts encoding pre- and postsynaptic proteins and transcripts implicated in autism spectrum disorders (ASD). We developed a brain polyribosome-programmed translation system, revealing that FMRP reversibly stalls ribosomes specifically on its target mRNAs. Our results suggest that loss of a translational brake on the synthesis of a subset of synaptic proteins contributes to FXS. In addition, they provide insight into the molecular basis of the cognitive and allied defects in FXS and ASD and suggest multiple targets for clinical intervention.


Subject(s)
Autistic Disorder/metabolism , Brain/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Ribosomes/metabolism , Synapses/metabolism , Animals , Autistic Disorder/physiopathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/physiopathology , Humans , Mice , Mice, Knockout , Polyribosomes/metabolism , Protein Biosynthesis , RNA-Binding Proteins , Sequence Analysis, RNA
7.
Proc Natl Acad Sci U S A ; 120(27): e2302534120, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37364131

ABSTRACT

Aberrant alternative splicing of mRNAs results in dysregulated gene expression in multiple neurological disorders. Here, we show that hundreds of mRNAs are incorrectly expressed and spliced in white blood cells and brain tissues of individuals with fragile X syndrome (FXS). Surprisingly, the FMR1 (Fragile X Messenger Ribonucleoprotein 1) gene is transcribed in >70% of the FXS tissues. In all FMR1-expressing FXS tissues, FMR1 RNA itself is mis-spliced in a CGG expansion-dependent manner to generate the little-known FMR1-217 RNA isoform, which is comprised of FMR1 exon 1 and a pseudo-exon in intron 1. FMR1-217 is also expressed in FXS premutation carrier-derived skin fibroblasts and brain tissues. We show that in cells aberrantly expressing mis-spliced FMR1, antisense oligonucleotide (ASO) treatment reduces FMR1-217, rescues full-length FMR1 RNA, and restores FMRP (Fragile X Messenger RibonucleoProtein) to normal levels. Notably, FMR1 gene reactivation in transcriptionally silent FXS cells using 5-aza-2'-deoxycytidine (5-AzadC), which prevents DNA methylation, increases FMR1-217 RNA levels but not FMRP. ASO treatment of cells prior to 5-AzadC application rescues full-length FMR1 expression and restores FMRP. These findings indicate that misregulated RNA-processing events in blood could serve as potent biomarkers for FXS and that in those individuals expressing FMR1-217, ASO treatment may offer a therapeutic approach to mitigate the disorder.


Subject(s)
Fragile X Syndrome , Humans , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Trinucleotide Repeat Expansion/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Decitabine , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Oligonucleotides , RNA
8.
J Neurochem ; 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225196

ABSTRACT

Perineuronal nets (PNNs) are mesh-like structures on the surfaces of parvalbumin-expressing inhibitory and other neurons, and consist of proteoglycans such as aggrecan, brevican, and neurocan. PNNs regulate the Excitatory/Inhibitory (E/I) balance in the brain and are formed at the closure of critical periods of plasticity during development. PNN formation is disrupted in Fragile X Syndrome, which is caused by silencing of the fragile X messenger ribonucleoprotein 1 (Fmr1) gene and loss of its protein product FMRP. FXS is characterized by impaired synaptic plasticity resulting in neuronal hyperexcitability and E/I imbalance. Here, we investigate how PNN formation is altered in FXS. PNNs are reduced in Fmr1 KO mouse brain when examined by staining for the lectin Wisteria floribunda agglutin (WFA) and aggrecan. Examination of PNNs by WFA staining at P14 and P42 in the hippocampus, somatosensory cortex, and retrosplenial cortex shows that they were reduced in these brain regions at P14 but mostly less so at P42 in Fmr1 KO mice. However, some differential FMRP regulation of PNN development in these brain regions persists, perhaps caused by asynchrony in PNN development between brain regions in wild-type animals. During development, aggrecan PNN levels in the brain were reduced in all brain regions in Fmr1 KO mice. Aggrecan mRNA levels were unchanged at these times, suggesting that FMRP is normally an activator of aggrecan mRNA translation. This hypothesis is buttressed by the observations that FMRP binds aggrecan mRNA and that ribosome profiling data show that aggrecan mRNA is associated with reduced numbers of ribosomes in Fmr1 KO mouse brain, indicating reduced translational efficiency. Moreover, aggrecan mRNA poly(A) tail length is also reduced in Fmr1 KO mouse brain, suggesting a relationship between polyadenylation and translational control. We propose a model where FMRP modulates PNN formation through translational up-regulation of aggrecan mRNA polyadenylation and translation.

9.
RNA ; 28(5): 756-765, 2022 05.
Article in English | MEDLINE | ID: mdl-35217597

ABSTRACT

Poly(A) tail length is regulated in both the nucleus and cytoplasm. One factor that controls polyadenylation in the cytoplasm is CPEB1, an RNA binding protein that associates with specific mRNA 3'UTR sequences to tether enzymes that add and remove poly(A). Two of these enzymes, the noncanonical poly(A) polymerases GLD2 (TENT2, PAPD4, Wispy) and GLD4 (TENT4B, PAPD5, TRF4, TUT3), interact with CPEB1 to extend poly(A). To identify additional RNA binding proteins that might anchor GLD4 to RNA, we expressed double tagged GLD4 in U87MG cells, which was used for sequential immunoprecipitation and elution followed by mass spectrometry. We identified several RNA binding proteins that coprecipitated with GLD4, among which was FMRP. To assess whether FMRP regulates polyadenylation, we performed TAIL-seq from WT and FMRP-deficient HEK293 cells. Surprisingly, loss of FMRP resulted in an overall increase in poly(A), which was also observed for several specific mRNAs. Conversely, loss of CPEB1 elicited an expected decrease in poly(A), which was examined in cultured neurons. We also examined polyadenylation in wild type (WT) and FMRP-deficient mouse brain cortex by direct RNA nanopore sequencing, which identified RNAs with both increased and decreased poly(A). Our data show that FMRP has a role in mediating poly(A) tail length, which adds to its repertoire of RNA regulation.


Subject(s)
Polyadenylation , mRNA Cleavage and Polyadenylation Factors , Animals , HEK293 Cells , Humans , Mice , Poly A/genetics , Poly A/metabolism , Polynucleotide Adenylyltransferase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
10.
RNA ; 28(2): 123-138, 2022 02.
Article in English | MEDLINE | ID: mdl-34848561

ABSTRACT

GGGGCC (G4C2) repeat expansion in the first intron of C9ORF72 causes amyotrophic lateral sclerosis and frontotemporal dementia. Repeat-containing RNA is translated into dipeptide repeat (DPR) proteins, some of which are neurotoxic. Using dynamic ribosome profiling, we identified three translation initiation sites in the intron upstream of (G4C2) repeats; these sites are detected irrespective of the presence or absence of the repeats. During translocation, ribosomes appear to be stalled on the repeats. An AUG in the preceding C9ORF72 exon initiates a uORF that inhibits downstream translation. Polysome isolation indicates that unspliced (G4C2) repeat-containing RNA is a substrate for DPR protein synthesis. (G4C2) repeat-containing RNA translation is 5' cap-independent but inhibited by the initiation factor DAP5, suggesting an interplay with uORF function. These results define novel translational mechanisms of expanded (G4C2) repeat-containing RNA in disease.


Subject(s)
C9orf72 Protein/genetics , Peptide Chain Initiation, Translational , RNA, Messenger/chemistry , Ribosomes/metabolism , C9orf72 Protein/metabolism , Dinucleotide Repeats , HEK293 Cells , HeLa Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Mol Psychiatry ; 28(7): 2728-2736, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37131078

ABSTRACT

The late 1990s were banner years in molecular neuroscience; seminal studies demonstrated that local protein synthesis, at or near synapses, was necessary for synaptic plasticity, the underlying cellular basis of learning and memory [1, 2]. The newly made proteins were proposed to "tag" the stimulated synapse, distinguishing it from naive synapses, thereby forming a cellular memory [3]. Subsequent studies demonstrated that the transport of mRNAs from soma to dendrite was linked with translational unmasking at synapses upon synaptic stimulation. It soon became apparent that one prevalent mechanism governing these events is cytoplasmic polyadenylation, and that among the proteins that control this process, CPEB, plays a central role in synaptic plasticity, and learning and memory. In vertebrates, CPEB is a family of four proteins, all of which regulate translation in the brain, that have partially overlapping functions, but also have unique characteristics and RNA binding properties that make them control different aspects of higher cognitive function. Biochemical analysis of the vertebrate CPEBs demonstrate them to respond to different signaling pathways whose output leads to specific cellular responses. In addition, the different CPEBs, when their functions go awry, result in pathophysiological phenotypes resembling specific human neurological disorders. In this essay, we review key aspects of the vertebrate CPEB proteins and cytoplasmic polyadenylation within the context of brain function.


Subject(s)
Polyadenylation , Transcription Factors , Animals , Humans , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism , Protein Biosynthesis , Neuronal Plasticity/physiology
12.
Cell ; 132(3): 335-7, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18267064

ABSTRACT

The translation of many maternal mRNAs is regulated by dynamic changes in poly(A) tail length. During maturation of Xenopus oocytes, polyadenylation is mediated by three different cis elements in the 3' untranslated region (UTR) of maternal mRNAs. In this issue, Piqué et al. (2008) explore the interplay of these elements to elucidate a combinatorial code that predicts the timing of polyadenylation and translation of maternal mRNAs.


Subject(s)
Polyadenylation , Protein Biosynthesis , RNA 3' Polyadenylation Signals , RNA, Messenger/metabolism , Animals , Models, Biological , Oocytes/metabolism , Xenopus laevis , mRNA Cleavage and Polyadenylation Factors/metabolism
13.
Proc Natl Acad Sci U S A ; 117(48): 30400-30411, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33199649

ABSTRACT

Fragile X syndrome (FXS) is caused by inactivation of the FMR1 gene and loss of encoded FMRP, an RNA binding protein that represses translation of some of its target transcripts. Here we use ribosome profiling and RNA sequencing to investigate the dysregulation of translation in the mouse brain cortex. We find that most changes in ribosome occupancy on hundreds of mRNAs are largely driven by dysregulation in transcript abundance. Many down-regulated mRNAs, which are mostly responsible for neuronal and synaptic functions, are highly enriched for FMRP binding targets. RNA metabolic labeling demonstrates that, in FMRP-deficient cortical neurons, mRNA down-regulation is caused by elevated degradation and is correlated with codon optimality. Moreover, FMRP preferentially binds mRNAs with optimal codons, suggesting that it stabilizes such transcripts through direct interactions via the translational machinery. Finally, we show that the paradigm of genetic rescue of FXS-like phenotypes in FMRP-deficient mice by deletion of the Cpeb1 gene is mediated by restoration of steady-state RNA levels and consequent rebalancing of translational homeostasis. Our data establish an essential role of FMRP in codon optimality-dependent mRNA stability as an important factor in FXS.


Subject(s)
Codon , Fragile X Mental Retardation Protein/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Cerebral Cortex/metabolism , Fragile X Syndrome/etiology , Fragile X Syndrome/metabolism , Gene Expression Profiling , Homeostasis , Mice , Models, Biological , Protein Biosynthesis , RNA Stability , Ribosomes/metabolism
14.
Glia ; 70(10): 1850-1863, 2022 10.
Article in English | MEDLINE | ID: mdl-35635122

ABSTRACT

Microglia are myeloid cells of the central nervous system that perform tasks essential for brain development, neural circuit homeostasis, and neural disease. Microglia react to inflammatory stimuli by upregulating inflammatory signaling through several different immune cell receptors such as the Toll-like receptor 4 (TLR4), which signals to several downstream effectors including transforming growth factor beta-activated kinase 1 (TAK1). Here, we show that TAK1 levels are regulated by CPEB1, a sequence-specific RNA binding protein that controls translation as well as RNA splicing and alternative poly(A) site selection in microglia. Lipopolysaccharide (LPS) binds the TLR4 receptor, which in CPEB1-deficient mice leads to elevated expression of ionized calcium binding adaptor molecule 1 (Iba1), a microglial protein that increases with inflammation, and increased levels of the cytokine IL6. This LPS-induced IL6 response is blocked by inhibitors of JNK, p38, ERK, NFκB, and TAK1. In contrast, phagocytosis, which is elevated in CPEB1-deficient microglia, is unaffected by LPS treatment or ERK inhibition, but is blocked by TAK1 inhibition. These data indicate that CPEB1 regulates microglial inflammatory responses and phagocytosis. RNA-seq indicates that these changes in inflammation and phagocytosis are accompanied by changes in RNA levels, splicing, and alternative poly(A) site selection. Thus, CPEB1 regulation of RNA expression plays a role in microglial function.


Subject(s)
Microglia , Phagocytosis , Polyadenylation , Transcription Factors , mRNA Cleavage and Polyadenylation Factors , Animals , Inflammation/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Mice , Microglia/metabolism , RNA/metabolism , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
15.
RNA Biol ; 18(7): 962-971, 2021 07.
Article in English | MEDLINE | ID: mdl-32954964

ABSTRACT

Noncanonical poly(A) polymerases are frequently tethered to mRNA 3' untranslated regions and regulate poly(A) tail length and resulting translation. In the brain, one such poly(A) polymerase is Gld2, which is anchored to mRNA by the RNA-binding protein CPEB1 to control local translation at postsynaptic regions. Depletion of CPEB1 or Gld2 from the mouse hippocampus results in a deficit in long-term potentiation (LTP), but only depletion of CPEB1 alters animal behaviour. To test whether a related enzyme, Gld4, compensates for the lack of Gld2, we separately or simultaneously depleted both proteins from hippocampal area CA1 and again found little change in animal behaviour, but observed a deficit in LTP as well as an increase in long-term depression (LTD), two forms of protein synthesis-dependent synaptic plasticity. RNA-seq data from Gld2, Gld4, and Gld2/Gld4-depleted hippocampus show widespread changes in steady state RNA levels, alternative splicing, and alternative poly(A) site selection. Many of the RNAs subject to these alterations encode proteins that mediate synaptic function, suggesting a molecular foundation for impaired synaptic plasticity.


Subject(s)
CA1 Region, Hippocampal/metabolism , Long-Term Potentiation/genetics , Polynucleotide Adenylyltransferase/genetics , RNA Processing, Post-Transcriptional , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , 3' Untranslated Regions , Animals , Avoidance Learning/physiology , CA1 Region, Hippocampal/pathology , Gene Expression Regulation , Injections, Intraventricular , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Obsessive Behavior/genetics , Obsessive Behavior/metabolism , Obsessive Behavior/physiopathology , Polynucleotide Adenylyltransferase/antagonists & inhibitors , Polynucleotide Adenylyltransferase/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic , mRNA Cleavage and Polyadenylation Factors/antagonists & inhibitors , mRNA Cleavage and Polyadenylation Factors/metabolism
16.
Nucleic Acids Res ; 47(5): e25, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30590705

ABSTRACT

Dysregulated protein synthesis is a major underlying cause of many neurodevelopmental diseases including fragile X syndrome. In order to capture subtle but biologically significant differences in translation in these disorders, a robust technique is required. One powerful tool to study translational control is ribosome profiling, which is based on deep sequencing of mRNA fragments protected from ribonuclease (RNase) digestion by ribosomes. However, this approach has been mainly applied to rapidly dividing cells where translation is active and large amounts of starting material are readily available. The application of ribosome profiling to low-input brain tissue where translation is modest and gene expression changes between genotypes are expected to be small has not been carefully evaluated. Using hippocampal tissue from wide type and fragile X mental retardation 1 (Fmr1) knockout mice, we show that variable RNase digestion can lead to significant sample batch effects. We also establish GC content and ribosome footprint length as quality control metrics for RNase digestion. We performed RNase titration experiments for low-input samples to identify optimal conditions for this critical step that is often improperly conducted. Our data reveal that optimal RNase digestion is essential to ensure high quality and reproducibility of ribosome profiling for low-input brain tissue.


Subject(s)
Brain/metabolism , Disease Models, Animal , Fragile X Syndrome/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Ribosomes/genetics , Ribosomes/metabolism , Animals , Base Sequence , Female , Fragile X Syndrome/metabolism , GC Rich Sequence , Male , Mice , Quality Control , RNA, Messenger/metabolism , Ribonucleases/metabolism
17.
Proc Natl Acad Sci U S A ; 115(48): E11397-E11405, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30373821

ABSTRACT

Fragile X syndrome (FXS) is caused by the loss of fragile X mental retardation protein (FMRP), an RNA binding protein whose deficiency impacts many brain functions, including differentiation of adult neural stem cells (aNSCs). However, the mechanism by which FMRP influences these processes remains unclear. Here, we performed ribosome profiling and transcriptomic analysis of aNSCs in parallel from wild-type and Fmr1 knockout mice. Our data revealed diverse gene expression changes at both mRNA and translation levels. Many mitosis and neurogenesis genes were dysregulated primarily at the mRNA level, while numerous synaptic genes were mostly dysregulated at the translation level. Translational "buffering", whereby changes in ribosome association with mRNA are compensated by alterations in RNA abundance, was also evident. Knockdown of NECDIN, an FMRP-repressed transcriptional factor, rescued neuronal differentiation. In addition, we discovered that FMRP regulates mitochondrial mRNA expression and energy homeostasis. Thus, FMRP controls diverse transcriptional and posttranscriptional gene expression programs critical for neural differentiation.


Subject(s)
Adult Stem Cells/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Neural Stem Cells/metabolism , Neurogenesis , RNA, Messenger/genetics , RNA, Mitochondrial/genetics , Adult Stem Cells/cytology , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Humans , Male , Mice , Mice, Knockout , Neural Stem Cells/cytology , RNA, Messenger/metabolism , RNA, Mitochondrial/metabolism
18.
Nat Rev Neurosci ; 16(10): 595-605, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26350240

ABSTRACT

Fragile X syndrome (FXS), the most-frequently inherited form of intellectual disability and the most-prevalent single-gene cause of autism, results from a lack of fragile X mental retardation protein (FMRP), an RNA-binding protein that acts, in most cases, to repress translation. Multiple pharmacological and genetic manipulations that target receptors, scaffolding proteins, kinases and translational control proteins can rescue neuronal morphology, synaptic function and behavioural phenotypes in FXS model mice, presumably by reducing excessive neuronal translation to normal levels. Such rescue strategies might also be explored in the future to identify the mRNAs that are critical for FXS pathophysiology.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Homeostasis/genetics , Animals , Fragile X Mental Retardation Protein/biosynthesis , Fragile X Syndrome/physiopathology , Gene Expression , Humans , Mice , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
19.
Mol Cell ; 47(2): 253-66, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22727665

ABSTRACT

Translational control of mRNAs in dendrites is essential for certain forms of synaptic plasticity and learning and memory. CPEB is an RNA-binding protein that regulates local translation in dendrites. Here, we identify poly(A) polymerase Gld2, deadenylase PARN, and translation inhibitory factor neuroguidin (Ngd) as components of a dendritic CPEB-associated polyadenylation apparatus. Synaptic stimulation induces phosphorylation of CPEB, PARN expulsion from the ribonucleoprotein complex, and polyadenylation in dendrites. A screen for mRNAs whose polyadenylation is altered by Gld2 depletion identified >100 transcripts including one encoding NR2A, an NMDA receptor subunit. shRNA depletion studies demonstrate that Gld2 promotes and Ngd inhibits dendritic NR2A expression. Finally, shRNA-mediated depletion of Gld2 in vivo attenuates protein synthesis-dependent long-term potentiation (LTP) at hippocampal dentate gyrus synapses; conversely, Ngd depletion enhances LTP. These results identify a pivotal role for polyadenylation and the opposing effects of Gld2 and Ngd in hippocampal synaptic plasticity.


Subject(s)
Cytoplasm/metabolism , Neuronal Plasticity , Protein Biosynthesis , Synaptic Transmission , Animals , Dendrites/metabolism , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Polyadenylation , Polynucleotide Adenylyltransferase/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/metabolism , Ribonucleoproteins/metabolism , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
20.
Nucleic Acids Res ; 45(11): 6793-6804, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28383716

ABSTRACT

Regulation of gene expression at the level of cytoplasmic polyadenylation is important for many biological phenomena including cell cycle progression, mitochondrial respiration, and learning and memory. GLD4 is one of the non-canonical poly(A) polymerases that regulates cytoplasmic polyadenylation-induced translation, but its target mRNAs and role in cellular physiology is not well known. To assess the full panoply of mRNAs whose polyadenylation is controlled by GLD4, we performed an unbiased whole genome-wide screen using poy(U) chromatography and thermal elution. We identified hundreds of mRNAs regulated by GLD4, several of which are involved in carbohydrate metabolism including GLUT1, a major glucose transporter. Depletion of GLD4 not only reduced GLUT1 poly(A) tail length, but also GLUT1 protein. GLD4-mediated translational control of GLUT1 mRNA is dependent of an RNA binding protein, CPEB1, and its binding elements in the 3΄ UTR. Through regulating GLUT1 level, GLD4 affects glucose uptake into cells and lactate levels. Moreover, GLD4 depletion impairs glucose deprivation-induced GLUT1 up-regulation. In addition, we found that GLD4 affects glucose-dependent cellular phenotypes such as migration and invasion in glioblastoma cells. Our observations delineate a novel post-transcriptional regulatory network involving carbohydrate metabolism and glucose homeostasis mediated by GLD4.


Subject(s)
Carbohydrate Metabolism , Polyadenylation , RNA Nucleotidyltransferases/physiology , Base Sequence , Cell Line, Tumor , Cell Movement , Cytoplasm/metabolism , Gene Expression Regulation , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , HEK293 Cells , Homeostasis , Humans , Protein Biosynthesis , RNA, Messenger/metabolism , Transcription Factors/physiology , mRNA Cleavage and Polyadenylation Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL