Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Science ; 377(6607): eabj6647, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35951695

ABSTRACT

Postmeiotic spermatids use a unique strategy to coordinate gene expression with morphological transformation, in which transcription and translation take place at separate developmental stages, but how mRNAs stored as translationally inert messenger ribonucleoproteins in developing spermatids become activated remains largely unknown. Here, we report that the RNA binding protein FXR1, a member of the fragile X-related (FXR) family, is highly expressed in late spermatids and undergoes liquid-liquid phase separation (LLPS) to merge messenger ribonucleoprotein granules with the translation machinery to convert stored mRNAs into a translationally activated state. Germline-specific Fxr1 ablation in mice impaired the translation of target mRNAs and caused defective spermatid development and male infertility, and a phase separation-deficient FXR1L351P mutation in Fxr1 knock-in mice produced the same developmental defect. These findings uncover a mechanism for translational reprogramming with LLPS as a key driver in spermiogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Protein Biosynthesis , RNA, Messenger, Stored , RNA-Binding Proteins , Spermatids , Spermatogenesis , Animals , Infertility, Male/genetics , Male , Mice , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spermatids/growth & development , Spermatids/metabolism , Spermatogenesis/genetics
2.
Nat Commun ; 10(1): 797, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30770808

ABSTRACT

FXR1 is an alternatively spliced gene that encodes RNA binding proteins (FXR1P) involved in muscle development. In contrast to other tissues, cardiac and skeletal muscle express two FXR1P isoforms that incorporate an additional exon-15. We report that recessive mutations in this particular exon of FXR1 cause congenital multi-minicore myopathy in humans and mice. Additionally, we show that while Myf5-dependent depletion of all FXR1P isoforms is neonatal lethal, mice carrying mutations in exon-15 display non-lethal myopathies which vary in severity depending on the specific effect of each mutation on the protein.


Subject(s)
Genes, Recessive , Genetic Predisposition to Disease/genetics , Muscle, Skeletal/metabolism , Mutation , Myopathies, Structural, Congenital/genetics , Ophthalmoplegia/genetics , RNA-Binding Proteins/genetics , Ryanodine Receptor Calcium Release Channel/deficiency , Animals , Cells, Cultured , Exons/genetics , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Mice, Transgenic , Myopathies, Structural, Congenital/congenital , Myopathies, Structural, Congenital/metabolism , Ophthalmoplegia/congenital , Ophthalmoplegia/metabolism , RNA-Binding Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism
3.
Cell Rep ; 9(4): 1402-1416, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456134

ABSTRACT

Translational control of mRNAs allows for rapid and selective changes in synaptic protein expression that are required for long-lasting plasticity and memory formation in the brain. Fragile X Related Protein 1 (FXR1P) is an RNA-binding protein that controls mRNA translation in nonneuronal cells and colocalizes with translational machinery in neurons. However, its neuronal mRNA targets and role in the brain are unknown. Here, we demonstrate that removal of FXR1P from the forebrain of postnatal mice selectively enhances long-term storage of spatial memories, hippocampal late-phase long-term potentiation (L-LTP), and de novo GluA2 synthesis. Furthermore, FXR1P binds specifically to the 5' UTR of GluA2 mRNA to repress translation and limit the amount of GluA2 that is incorporated at potentiated synapses. This study uncovers a mechanism for regulating long-lasting synaptic plasticity and spatial memory formation and reveals an unexpected divergent role of FXR1P among Fragile X proteins in brain plasticity.


Subject(s)
Long-Term Potentiation , Memory, Long-Term , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Receptors, AMPA/biosynthesis , Synapses/metabolism , 5' Untranslated Regions/genetics , Animals , Behavior, Animal , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
J Neurosci ; 31(39): 13705-9, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21957233

ABSTRACT

Mammalian brain-specific miR-9 and miR-124 have been implicated in several aspects of neuronal development and function. However, it is not known how their expression levels are regulated in vivo. We found that the levels of miR-9 and miR-124 are regulated by FXR1P but not by the loss of FXR2P or FMRP in vivo, a mouse model of fragile X syndrome. Surprisingly, the levels of miR-9 and miR-124 are elevated in fmr1/fxr2 double-knock-out mice, in part reflecting posttranscriptional upregulation of FXR1P. Indeed, FXR1P is required for efficient processing of pre-miR-9 and pre-miR-124 in vitro and forms a complex with Dicer and pre-miRNAs. These findings reveal differential roles of FMRP family proteins in controlling the expression levels of brain-specific miRNAs.


Subject(s)
Brain/metabolism , Fragile X Mental Retardation Protein , MicroRNAs/metabolism , RNA-Binding Proteins/physiology , Animals , Cells, Cultured , DEAD-box RNA Helicases/metabolism , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , MicroRNAs/biosynthesis , RNA-Binding Proteins/genetics , Ribonuclease III/metabolism
5.
Nat Med ; 17(5): 559-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21516088

ABSTRACT

Deficiency in fragile X mental retardation protein (FMRP) results in fragile X syndrome (FXS), an inherited form of intellectual disability. Despite extensive research, it is unclear how FMRP deficiency contributes to the cognitive deficits in FXS. Fmrp-null mice show reduced adult hippocampal neurogenesis. As Fmrp is also enriched in mature neurons, we investigated the function of Fmrp expression in neural stem and progenitor cells (aNSCs) and its role in adult neurogenesis. Here we show that ablation of Fmrp in aNSCs by inducible gene recombination leads to reduced hippocampal neurogenesis in vitro and in vivo, as well as markedly impairing hippocampus-dependent learning in mice. Conversely, restoration of Fmrp expression specifically in aNSCs rescues these learning deficits in Fmrp-deficient mice. These data suggest that defective adult neurogenesis may contribute to the learning impairment seen in FXS, and these learning deficits can be rectified by delayed restoration of Fmrp specifically in aNSCs.


Subject(s)
Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/physiopathology , Hippocampus/physiopathology , Learning/physiology , Adult Stem Cells/physiology , Animals , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Fragile X Syndrome/psychology , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/physiology , Neurogenesis/genetics , Neurogenesis/physiology
6.
Mol Cancer ; 6: 23, 2007 Mar 26.
Article in English | MEDLINE | ID: mdl-17386101

ABSTRACT

Bright/ARID3A is a nuclear matrix-associated transcription factor that stimulates immunoglobulin heavy chain (IgH) expression and Cyclin E1/E2F-dependent cell cycle progression. Bright positively activates IgH transcriptional initiation by binding to ATC-rich P sites within nuclear matrix attachment regions (MARs) flanking the IgH intronic enhancer (Emu). Over-expression of Bright in cultured B cells was shown to correlate with DNase hypersensitivity of Emu. We report here further efforts to analyze Bright-mediated Emu enhancer activation within the physiological constraints of chromatin. A system was established in which VH promoter-driven in vitro transcription on chromatin- reconstituted templates was responsive to Emu. Bright assisted in blocking the general repression caused by nucleosome assembly but was incapable of stimulating transcription from prebound nucleosome arrays. In vitro transcriptional derepression by Bright was enhanced on templates in which Emu is flanked by MARs and was inhibited by competition with high affinity Bright binding (P2) sites. DNase hypersensitivity of chromatin-reconstituted Emu was increased when prepackaged with B cell nuclear extract supplemented with Bright. These results identify Bright as a contributor to accessibility of the IgH enhancer.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Immunoglobulin Heavy Chains/genetics , Trans-Activators/metabolism , Cell Line , Deoxyribonucleases/metabolism , Humans , Oncogenes , Promoter Regions, Genetic/genetics , Protein Binding , Transcription Factors , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...