Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Hum Mol Genet ; 31(9): 1453-1470, 2022 05 04.
Article in English | MEDLINE | ID: mdl-34791230

ABSTRACT

Spinal muscular atrophy (SMA) is characterized by the loss of alpha motor neurons in the spinal cord and a progressive muscle weakness and atrophy. SMA is caused by loss-of-function mutations and/or deletions in the survival of motor neuron (SMN) gene. The role of SMN in motor neurons has been extensively studied, but its function and the consequences of its loss in muscle have also emerged as a key aspect of SMA pathology. In this study, we explore the molecular mechanisms involved in muscle defects in SMA. First, we show in C2C12 myoblasts, that arginine methylation by CARM1 controls myogenic differentiation. More specifically, the methylation of HuR on K217 regulates HuR levels and subcellular localization during myogenic differentiation, and the formation of myotubes. Furthermore, we demonstrate that SMN and HuR interact in C2C12 myoblasts. Interestingly, the SMA-causing E134K point mutation within the SMN Tudor domain, and CARM1 depletion, modulate the SMN-HuR interaction. In addition, using the Smn2B/- mouse model, we report that CARM1 levels are markedly increased in SMA muscles and that HuR fails to properly respond to muscle denervation, thereby affecting the regulation of its mRNA targets. Altogether, our results show a novel CARM1-HuR axis in the regulation of muscle differentiation and plasticity as well as in the aberrant regulation of this axis caused by the absence of SMN in SMA muscle. With the recent developments of therapeutics targeting motor neurons, this study further indicates the need for more global therapeutic approaches for SMA.


Subject(s)
Muscular Atrophy, Spinal , Animals , Disease Models, Animal , ELAV-Like Protein 1 , Mice , Motor Neurons/metabolism , Muscles/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Survival of Motor Neuron 1 Protein/genetics
2.
Cell Mol Life Sci ; 80(11): 328, 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37847286

ABSTRACT

Elevated mitochondrial metabolism promotes tumorigenesis of Embryonal Rhabdomyosarcomas (ERMS). Accordingly, targeting oxidative phosphorylation (OXPHOS) could represent a therapeutic strategy for ERMS. We previously demonstrated that genetic reduction of Staufen1 (STAU1) levels results in the inhibition of ERMS tumorigenicity. Here, we examined STAU1-mediated mechanisms in ERMS and focused on its potential involvement in regulating OXPHOS. We report the novel and differential role of STAU1 in mitochondrial metabolism in cancerous versus non-malignant skeletal muscle cells (NMSkMCs). Specifically, our data show that STAU1 depletion reduces OXPHOS and inhibits proliferation of ERMS cells. Our findings further reveal the binding of STAU1 to several OXPHOS mRNAs which affects their stability. Indeed, STAU1 depletion reduced the stability of OXPHOS mRNAs, causing inhibition of mitochondrial metabolism. In parallel, STAU1 depletion impacted negatively the HIF2α pathway which further modulates mitochondrial metabolism. Exogenous expression of HIF2α in STAU1-depleted cells reversed the mitochondrial inhibition and induced cell proliferation. However, opposite effects were observed in NMSkMCs. Altogether, these findings revealed the impact of STAU1 in the regulation of mitochondrial OXPHOS in cancer cells as well as its differential role in NMSkMCs. Overall, our results highlight the therapeutic potential of targeting STAU1 as a novel approach for inhibiting mitochondrial metabolism in ERMS.


Subject(s)
Rhabdomyosarcoma, Embryonal , Humans , Rhabdomyosarcoma, Embryonal/genetics , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/metabolism , Cytoskeletal Proteins/metabolism , Cell Transformation, Neoplastic , Carcinogenesis/genetics , Cell Proliferation/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism
3.
Methods ; 175: 44-52, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31794835

ABSTRACT

The protein arginine methyltransferase family (PRMT) is known as being the catalytic driving force for arginine methylation. This specific type of post translational modification is extensively used in biological processes, and therefore is highly relevant in the pathology of a profusion of diseases. Since altered PRMT expression or deregulation has been shown to contribute to a vast range of those diseases including cancer, their study is of great interest. Although an increasing number of substrates are being discovered for each PRMT, large scale proteomic methods can be used to identify novel interactors/substrates, further elucidating the role that PRMTs perform in physiological or disease states. Here, we describe the use of affinity purification (AP) coupled with stable isotope labeling with amino acids in cell culture (SILAC) quantitative mass spectrometry (MS) to identify protein interactors and substrates of PRMTs. We also explore the possibility of exploiting the fact most PRMTs display lower dissociation rates with their hypomethylated substrates as a strategy to increase the proportion of substrates identified in AP/MS studies.


Subject(s)
Chromatography, Affinity/methods , Enzyme Inhibitors/chemistry , Mass Spectrometry/methods , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/chemistry , Proteomics/methods , Amino Acids/metabolism , Arginine/analysis , Arginine/chemistry , Arginine/metabolism , Gene Expression , Histones/chemistry , Histones/metabolism , Humans , Isotope Labeling , MCF-7 Cells , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding/drug effects , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Recombinant Proteins
4.
PLoS Genet ; 13(2): e1006626, 2017 02.
Article in English | MEDLINE | ID: mdl-28231279

ABSTRACT

Eukaryotic cells form stress granules under a variety of stresses, however the signaling pathways regulating their formation remain largely unknown. We have determined that the Saccharomyces cerevisiae lysine acetyltransferase complex NuA4 is required for stress granule formation upon glucose deprivation but not heat stress. Further, the Tip60 complex, the human homolog of the NuA4 complex, is required for stress granule formation in cancer cell lines. Surprisingly, the impact of NuA4 on glucose-deprived stress granule formation is partially mediated through regulation of acetyl-CoA levels, which are elevated in NuA4 mutants. While elevated acetyl-CoA levels suppress the formation of glucose-deprived stress granules, decreased acetyl-CoA levels enhance stress granule formation upon glucose deprivation. Further our work suggests that NuA4 regulates acetyl-CoA levels through the Acetyl-CoA carboxylase Acc1. Altogether this work establishes both NuA4 and the metabolite acetyl-CoA as critical signaling pathways regulating the formation of glucose-deprived stress granules.


Subject(s)
Acetyl Coenzyme A/genetics , Acetyltransferases/genetics , Glucose/metabolism , Histone Acetyltransferases/genetics , Saccharomyces cerevisiae Proteins/genetics , Histone Acetyltransferases/biosynthesis , Histone Acetyltransferases/metabolism , Humans , Lysine Acetyltransferase 5 , Mutant Proteins/biosynthesis , Mutant Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Stress, Physiological/genetics
5.
Hum Mol Genet ; 26(12): 2192-2206, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28369518

ABSTRACT

Myotonic Dystrophy type 1 (DM1) is caused by an expansion of CUG repeats in DMPK mRNAs. This mutation affects alternative splicing through misregulation of RNA-binding proteins. Amongst pre-mRNAs that are mis-spliced, several code for proteins involved in calcium homeostasis suggesting that calcium-handling and signaling are perturbed in DM1. Here, we analyzed expression of such proteins in DM1 mouse muscle. We found that the levels of several sarcoplasmic reticulum proteins (SERCA1, sarcolipin and calsequestrin) are altered, likely contributing to an imbalance in calcium homeostasis. We also observed that calcineurin (CnA) signaling is hyperactivated in DM1 muscle. Indeed, CnA expression and phosphatase activity are both markedly increased in DM1 muscle. Coherent with this, we found that activators of the CnA pathway (MLP, FHL1) are also elevated. Consequently, NFATc1 expression is increased in DM1 muscle and becomes relocalized to myonuclei, together with an up-regulation of its transcriptional targets (RCAN1.4 and myoglobin). Accordingly, DM1 mouse muscles display an increase in oxidative metabolism and fiber hypertrophy. To determine the functional consequences of this CnA hyperactivation, we administered cyclosporine A, an inhibitor of CnA, to DM1 mice. Muscles of treated DM1 mice showed an increase in CUGBP1 levels, and an exacerbation of key alternative splicing events associated with DM1. Finally, inhibition of CnA in cultured human DM1 myoblasts also resulted in a splicing exacerbation of the insulin receptor. Together, these findings show for the first time that calcium-CnA signaling is hyperactivated in DM1 muscle and that such hyperactivation represents a beneficial compensatory adaptation to the disease.


Subject(s)
Calcineurin/metabolism , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Alternative Splicing , Animals , Antigens, CD , Calcineurin/genetics , Calcium/metabolism , Calcium Signaling , Cell Culture Techniques , Disease Models, Animal , Fibroblasts/metabolism , Homeostasis , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/metabolism , NFATC Transcription Factors , RNA Splicing , RNA, Messenger/genetics , RNA-Binding Proteins , Receptor, Insulin , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Signal Transduction , Up-Regulation
6.
Hum Mol Genet ; 26(10): 1821-1838, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28369467

ABSTRACT

Converging lines of evidence have now highlighted the key role for post-transcriptional regulation in the neuromuscular system. In particular, several RNA-binding proteins are known to be misregulated in neuromuscular disorders including myotonic dystrophy type 1, spinal muscular atrophy and amyotrophic lateral sclerosis. In this study, we focused on the RNA-binding protein Staufen1, which assumes multiple functions in both skeletal muscle and neurons. Given our previous work that showed a marked increase in Staufen1 expression in various physiological and pathological conditions including denervated muscle, in embryonic and undifferentiated skeletal muscle, in rhabdomyosarcomas as well as in myotonic dystrophy type 1 muscle samples from both mouse models and humans, we investigated the impact of sustained Staufen1 expression in postnatal skeletal muscle. To this end, we generated a skeletal muscle-specific transgenic mouse model using the muscle creatine kinase promoter to drive tissue-specific expression of Staufen1. We report that sustained Staufen1 expression in postnatal skeletal muscle causes a myopathy characterized by significant morphological and functional deficits. These deficits are accompanied by a marked increase in the expression of several atrophy-associated genes and by the negative regulation of PI3K/AKT signaling. We also uncovered that Staufen1 mediates PTEN expression through indirect transcriptional and direct post-transcriptional events thereby providing the first evidence for Staufen1-regulated PTEN expression. Collectively, our data demonstrate that Staufen1 is a novel atrophy-associated gene, and highlight its potential as a biomarker and therapeutic target for neuromuscular disorders and conditions.


Subject(s)
RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Gene Expression , Mice , Mice, Knockout , Muscle Denervation , Muscle, Skeletal/metabolism , Muscles/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy, Spinal/metabolism , Myotonic Dystrophy/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , RNA/metabolism , RNA Processing, Post-Transcriptional , Signal Transduction , Tensins
7.
PLoS Genet ; 12(1): e1005827, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26824521

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a neuromuscular disorder caused by an expansion of CUG repeats in the 3' UTR of the DMPK gene. The CUG repeats form aggregates of mutant mRNA, which cause misregulation and/or sequestration of RNA-binding proteins, causing aberrant alternative splicing in cells. Previously, we showed that the multi-functional RNA-binding protein Staufen1 (Stau1) was increased in skeletal muscle of DM1 mouse models and patients. We also showed that Stau1 rescues the alternative splicing profile of pre-mRNAs, e.g. the INSR and CLC1, known to be aberrantly spliced in DM1. In order to explore further the potential of Stau1 as a therapeutic target for DM1, we first investigated the mechanism by which Stau1 regulates pre-mRNA alternative splicing. We report here that Stau1 regulates the alternative splicing of exon 11 of the human INSR via binding to Alu elements located in intron 10. Additionally, using a high-throughput RT-PCR screen, we have identified numerous Stau1-regulated alternative splicing events in both WT and DM1 myoblasts. A number of these aberrant ASEs in DM1, including INSR exon 11, are rescued by overexpression of Stau1. However, we find other ASEs in DM1 cells, where overexpression of Stau1 shifts the splicing patterns away from WT conditions. Moreover, we uncovered that Stau1-regulated ASEs harbour Alu elements in intronic regions flanking the alternative exon more than non-Stau1 targets. Taken together, these data highlight the broad impact of Stau1 as a splicing regulator and suggest that Stau1 may act as a disease modifier in DM1.


Subject(s)
Alternative Splicing/genetics , Cytoskeletal Proteins/genetics , Myotonin-Protein Kinase/genetics , RNA-Binding Proteins/genetics , Trinucleotide Repeat Expansion/genetics , 3' Untranslated Regions , Alu Elements/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cytoskeletal Proteins/metabolism , Humans , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/metabolism , Myoblasts/pathology , Myotonic Dystrophy , Myotonin-Protein Kinase/metabolism , Protein Binding , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
8.
Nucleic Acids Res ; 44(6): 2661-76, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26656492

ABSTRACT

Loss of 'Survival of Motor Neurons' (SMN) leads to spinal muscular atrophy (SMA), a disease characterized by degeneration of spinal cord alpha motor neurons, resulting in muscle weakness, paralysis and death during early childhood. SMN is required for assembly of the core splicing machinery, and splicing defects were documented in SMA. We previously uncovered that Coactivator-Associated Methyltransferase-1 (CARM1) is abnormally up-regulated in SMA, leading to mis-regulation of a number of transcriptional and alternative splicing events. We report here that CARM1 can promote decay of a premature terminating codon (PTC)-containing mRNA reporter, suggesting it can act as a mediator of nonsense-mediated mRNA decay (NMD). Interestingly, this pathway, while originally perceived as solely a surveillance mechanism preventing expression of potentially detrimental proteins, is now emerging as a highly regulated RNA decay pathway also acting on a subset of normal mRNAs. We further show that CARM1 associates with major NMD factor UPF1 and promotes its occupancy on PTC-containing transcripts. Finally, we identify a specific subset of NMD targets that are dependent on CARM1 for degradation and that are also misregulated in SMA, potentially adding exacerbated targeting of PTC-containing mRNAs to the already complex array of molecular defects associated with this disease.


Subject(s)
Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Nonsense Mediated mRNA Decay , Protein-Arginine N-Methyltransferases/genetics , RNA, Messenger/genetics , Trans-Activators/genetics , Alternative Splicing , Animals , Cell Line , Codon, Terminator , Exons , Humans , Introns , Mice , Mice, Inbred C57BL , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Protein-Arginine N-Methyltransferases/metabolism , RNA Helicases , RNA, Messenger/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Trans-Activators/metabolism
9.
J Neurosci ; 35(34): 12063-79, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311784

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by the selective loss of spinal motor neurons due to the depletion of the survival of motor neuron (SMN) protein. No therapy is currently available for SMA, which represents the leading genetic cause of death in childhood. In the present study, we report that insulin-like growth factor-1 receptor (Igf-1r) gene expression is enhanced in the spinal cords of SMA-like mice. The reduction of expression, either at the physiological (through physical exercise) or genetic level, resulted in the following: (1) a significant improvement in lifespan and motor behavior, (2) a significant motor neuron protection, and (3) an increase in SMN expression in spinal cord and skeletal muscles through both transcriptional and posttranscriptional mechanisms. Furthermore, we have found that reducing IGF-1R expression is sufficient to restore intracellular signaling pathway activation profile lying downstream of IGF-1R, resulting in both the powerful activation of the neuroprotective AKT/CREB pathway and the inhibition of the ERK and JAK pathways. Therefore, reducing rather than enhancing the IGF-1 pathway could constitute a useful strategy to limit neurodegeneration in SMA. SIGNIFICANCE STATEMENT: Recent evidence of IGF-1 axis alteration in spinal muscular atrophy (SMA), a very severe neurodegenerative disease affecting specifically the motor neurons, have triggered a renewed interest in insulin-like growth factor-1 (IGF-1) pathway activation as a potential therapeutic approach for motor neuron diseases. The present study challenges this point of view and brings the alternative hypothesis that reducing rather than enhancing the IGF-1 signaling pathway exerts a neuroprotective effect in SMA. Furthermore, the present data substantiate a newly emerging concept that the modulation of IGF-1 receptor expression is a key event selectively determining the activation level of intracellular pathways that lie downstream of the receptor. This aspect should be considered when designing IGF-1-based treatments for neurodegenerative diseases.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/prevention & control , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Receptor, IGF Type 1/genetics
10.
Nucleic Acids Res ; 42(6): 3982-97, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24371285

ABSTRACT

Several reports have previously highlighted the potential role of miR-206 in the post-transcriptional downregulation of utrophin A in cultured cells. Along those lines, we recently identified K-homology splicing regulator protein (KSRP) as an important negative regulator in the post-transcriptional control of utrophin A in skeletal muscle. We sought to determine whether these two pathways act together to downregulate utrophin A expression in skeletal muscle. Surprisingly, we discovered that miR-206 overexpression in cultured cells and dystrophic muscle fibers causes upregulation of endogenous utrophin A levels. We further show that this upregulation of utrophin A results from the binding of miR-206 to conserved sites located in the 3'-UTR (untranslated region) of KSRP, thus causing the subsequent inhibition of KSRP expression. This miR-206-mediated decrease in KSRP levels leads, in turn, to an increase in the expression of utrophin A due to a reduction in the activity of this destabilizing RNA-binding protein. Our work shows that miR-206 can oscillate between direct repression of utrophin A expression via its 3'-UTR and activation of its expression through decreased availability of KSRP and interactions with AU-rich elements located within the 3'-UTR of utrophin A. Our study thus reveals that two apparent negative post-transcriptional pathways can act distinctively as molecular switches causing repression or activation of utrophin A expression.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Utrophin/metabolism , 3' Untranslated Regions , Animals , Cell Differentiation , Cell Line , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/cytology , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Up-Regulation , Utrophin/genetics
11.
PLoS Genet ; 9(10): e1003890, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204304

ABSTRACT

Fragile X syndrome is caused by loss of function of a single gene encoding the Fragile X Mental Retardation Protein (FMRP). This RNA-binding protein, widely expressed in mammalian tissues, is particularly abundant in neurons and is a component of messenger ribonucleoprotein (mRNP) complexes present within the translational apparatus. The absence of FMRP in neurons is believed to cause translation dysregulation and defects in mRNA transport essential for local protein synthesis and for synaptic development and maturation. A prevalent model posits that FMRP is a nucleocytoplasmic shuttling protein that transports its mRNA targets from the nucleus to the translation machinery. However, it is not known which of the multiple FMRP isoforms, resulting from the numerous alternatively spliced FMR1 transcripts variants, would be involved in such a process. Using a new generation of anti-FMRP antibodies and recombinant expression, we show here that the most commonly expressed human FMRP isoforms (ISO1 and 7) do not localize to the nucleus. Instead, specific FMRP isoforms 6 and 12 (ISO6 and 12), containing a novel C-terminal domain, were the only isoforms that localized to the nuclei in cultured human cells. These isoforms localized to specific p80-coilin and SMN positive structures that were identified as Cajal bodies. The Cajal body localization signal was confined to a 17 amino acid stretch in the C-terminus of human ISO6 and is lacking in a mouse Iso6 variant. As FMRP is an RNA-binding protein, its presence in Cajal bodies suggests additional functions in nuclear post-transcriptional RNA metabolism. Supporting this hypothesis, a missense mutation (I304N), known to alter the KH2-mediated RNA binding properties of FMRP, abolishes the localization of human FMRP ISO6 to Cajal bodies. These findings open unexplored avenues in search for new insights into the pathophysiology of Fragile X Syndrome.


Subject(s)
Coiled Bodies/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Protein Isoforms/biosynthesis , Animals , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Coiled Bodies/ultrastructure , Fragile X Mental Retardation Protein/biosynthesis , Fragile X Syndrome/pathology , Gene Expression Regulation , Humans , Mice , Neurons/metabolism , Protein Isoforms/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribonucleoproteins/genetics
12.
Proteomics ; 15(13): 2187-97, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25690678

ABSTRACT

Arginine methylation is catalyzed by a family of enzymes called protein arginine methyltransferases (PRMTs). The PRMT1 gene generates at least seven distinct alternatively spliced isoforms (PRMT v1-v7), which together contribute a significant portion of the cellular arginine methylome. The distinct biochemical and biological functions of these PRMT1 isoforms have not been well characterized. Previously we have shown that while both PRMT1v1 and PRMT1v2 are overexpressed in breast cancer cells, PRMT1v2 specifically promotes breast cancer cell survival and invasion. These isoforms also have distinct subcellular localizations, PRMT1v1 is mainly nuclear and PRMT1v2 cytosolic. To gain further knowledge into their isoform-specific roles within cells we used a SILAC-based quantitative affinity purification/MS approach to identify their individual protein interactomes in breast cancer cells. This analysis has uncovered distinct interactomes for PRMT1v1 and PRMT1v2. Consistent with their distinct subcellular localizations, PRMT1v1 enriched a mainly nuclear protein interactome, while PRMT1v2 enriched predominantly cytoplasmic interactors from whole-cell extracts. Furthermore, these interactomes revealed that PRMT1v1 has a role in regulating gene expression, while PRMT1v2 functions in cytoskeletal dynamics. These results highlight the unique functions of these isoforms and the distinct roles they may play within cells, with potential implications for breast cancer and other diseases.


Subject(s)
Breast Neoplasms/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Mass Spectrometry , Microscopy, Fluorescence , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Hum Mol Genet ; 22(15): 3093-111, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23575223

ABSTRACT

Several therapeutic approaches are currently being developed for Duchenne muscular dystrophy (DMD) including upregulating the levels of endogenous utrophin A in dystrophic fibers. Here, we examined the role of post-transcriptional mechanisms in controlling utrophin A expression in skeletal muscle. We show that activation of p38 leads to an increase in utrophin A independently of a transcriptional induction. Rather, p38 controls the levels of utrophin A mRNA by extending the half-life of transcripts via AU-rich elements (AREs). This mechanism critically depends on a decrease in the functional availability of KSRP, an RNA-binding protein known to promote decay of ARE-containing transcripts. In vitro and in vivo binding studies revealed that KSRP interacts with specific AREs located within the utrophin A 3' UTR. Electroporation experiments to knockdown KSRP led to an increase in utrophin A in wild-type and mdx mouse muscles. In pre-clinical studies, treatment of mdx mice with heparin, an activator of p38, causes a pronounced increase in utrophin A in diaphragm muscle fibers. Together, these studies identify a pathway that culminates in the post-transcriptional regulation of utrophin A through increases in mRNA stability. Furthermore, our results constitute proof-of-principle showing that pharmacological activation of p38 may prove beneficial as a novel therapeutic approach for DMD.


Subject(s)
AU Rich Elements , Muscle, Skeletal/metabolism , RNA Stability , RNA-Binding Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Utrophin/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , 3' Untranslated Regions , Animals , Enzyme Activation , Gene Expression Regulation/drug effects , Heparin/pharmacology , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Protein Binding , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Utrophin/metabolism
14.
Hum Mol Genet ; 22(4): 668-84, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23136128

ABSTRACT

SMN1, the causative gene for spinal muscular atrophy (SMA), plays a housekeeping role in the biogenesis of small nuclear RNA ribonucleoproteins. SMN is also present in granular foci along axonal projections of motoneurons, which are the predominant cell type affected in the pathology. These so-called RNA granules mediate the transport of specific mRNAs along neurites and regulate mRNA localization, stability, as well as local translation. Recent work has provided evidence suggesting that SMN may participate in the assembly of RNA granules, but beyond that, the precise nature of its role within these structures remains unclear. Here, we demonstrate that SMN associates with polyribosomes and can repress translation in an in vitro translation system. We further identify the arginine methyltransferase CARM1 as an mRNA that is regulated at the translational level by SMN and find that CARM1 is abnormally up-regulated in spinal cord tissue from SMA mice and in severe type I SMA patient cells. We have previously characterized a novel regulatory pathway in motoneurons involving the SMN-interacting RNA-binding protein HuD and CARM1. Thus, our results suggest the existence of a potential negative feedback loop in this pathway. Importantly, an SMA-causing mutation in the Tudor domain of SMN completely abolished translational repression, a strong indication for the functional significance of this novel SMN activity in the pathology.


Subject(s)
Gene Expression Regulation, Enzymologic , Protein Biosynthesis , Survival of Motor Neuron 1 Protein/genetics , Animals , Cells, Cultured , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Polyribosomes/metabolism , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Spinal Cord/enzymology , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 1 Protein/physiology , Untranslated Regions , Up-Regulation
15.
Mutagenesis ; 30(2): 177-89, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25688111

ABSTRACT

Breast cancer is the most commonly diagnosed female cancer in the world. Though therapeutic treatments are available to treat breast cancer and in some instances are successful, the occurrence of unsuccessful treatment, or the rate of tumour recurrence, still remains strikingly high. Therefore, novel therapeutic treatment targets need to be discovered and tested. The protein arginine methyltransferases (PRMTs) are a family of enzymes that catalyse arginine methylation and are implicated in a myriad of cellular pathways including transcription, DNA repair, RNA metabolism, signal transduction, protein-protein interactions and subcellular localisation. In breast cancer, the expression levels and enzymatic activity of a number of PRMTs is dysregulated; significantly altering the regulation of many cellular pathways that are implicated in breast cancer development and progression. Here, we review the current knowledge on PRMTs in breast cancer and provide a rationale for how PRMTs may provide novel therapeutic targets for the treatment of breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Female , Humans , Protein-Arginine N-Methyltransferases/physiology
16.
Hum Mol Genet ; 20(3): 553-79, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21088113

ABSTRACT

Spinal muscular atrophy is an autosomal-recessive neuromuscular disease caused by disruption of the survival of motor neuron (SMN) gene, which promotes cytoplasmic assembly of the splicing core machinery. It remains unclear how a deficiency in SMN results in a disorder leading to selective degeneration of lower motor neurons. We report here that SMN interacts with RNA-binding protein HuD in neurites of motorneuron-derived MN-1 cells. This interaction is mediated through the Tudor domain of SMN and, importantly, naturally occurring Tudor mutations found in patients with severe spinal muscular atrophy (SMA) completely abrogate the interaction, underscoring its relevance to the disease process. We also characterized a regulatory pathway involving coactivator-associated arginine methyltransferase 1 (CARM1) and HuD. Specifically, we show that CARM1 expression is rapidly downregulated, at the protein level, following induction of differentiation through retinoid and neurotrophic signaling. Using purified proteins, we demonstrate that methylation of HuD by CARM1 reduces its interaction with the p21(cip1/waf1) mRNA, showing that CARM1 can directly influence RNA-binding activity. We further demonstrate that this CARM1-dependent regulatory switch mainly controls the activity of HuD in promoting cell-cycle exit, whereas the interaction between HuD and SMN is required for proper recruitment of HuD and its mRNA targets in neuronal RNA granules. Finally, we were able to rescue SMA-like defects in a hypomorphic Smn knockdown MN-1 cell line through overexpression of HuD. Together, these findings extend our understanding of specific role(s) of SMN in motor neurons and provide crucial insights into potential new avenues for SMA therapeutic strategies.


Subject(s)
ELAV Proteins/metabolism , Muscular Atrophy, Spinal/metabolism , Neurites/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Base Sequence , Cell Cycle , Cell Cycle Proteins/genetics , Cell Line , ELAV-Like Protein 4 , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Immunoblotting , Methylation , Mice , Muscular Atrophy, Spinal/genetics , Mutation , Nerve Growth Factors , Phenotype , Polymerase Chain Reaction , Protein Interaction Domains and Motifs , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Retinol-Binding Proteins , Signal Transduction
17.
Life Sci Alliance ; 6(6)2023 06.
Article in English | MEDLINE | ID: mdl-36882285

ABSTRACT

Spinal muscular atrophy is the leading genetic cause of infant mortality and results from depleted levels of functional survival of motor neuron (SMN) protein by either deletion or mutation of the SMN1 gene. SMN is characterized by a central TUDOR domain, which mediates the association of SMN with arginine methylated (Rme) partners, such as coilin, fibrillarin, and RNA pol II (RNA polymerase II). Herein, we biochemically demonstrate that SMN also associates with histone H3 monomethylated on lysine 79 (H3K79me1), defining SMN as not only the first protein known to associate with the H3K79me1 histone modification but also the first histone mark reader to recognize both methylated arginine and lysine residues. Mutational analyzes provide evidence that SMNTUDOR associates with H3 via an aromatic cage. Importantly, most SMNTUDOR mutants found in spinal muscular atrophy patients fail to associate with H3K79me1.


Subject(s)
Histone Code , Muscular Atrophy, Spinal , Survival of Motor Neuron 1 Protein , Humans , Infant , Arginine , Lysine , Muscular Atrophy, Spinal/genetics , RNA Polymerase II , Transcription Factors , Survival of Motor Neuron 1 Protein/genetics
18.
Life Sci Alliance ; 6(1)2023 01.
Article in English | MEDLINE | ID: mdl-36375840

ABSTRACT

Although recent advances in gene therapy provide hope for spinal muscular atrophy (SMA) patients, the pathology remains the leading genetic cause of infant mortality. SMA is a monogenic pathology that originates from the loss of the SMN1 gene in most cases or mutations in rare cases. Interestingly, several SMN1 mutations occur within the TUDOR methylarginine reader domain of SMN. We hypothesized that in SMN1 mutant cases, SMA may emerge from aberrant protein-protein interactions between SMN and key neuronal factors. Using a BioID proteomic approach, we have identified and validated a number of SMN-interacting proteins, including fragile X mental retardation protein (FMRP) family members (FMRFM). Importantly, SMA-linked SMNTUDOR mutant forms (SMNST) failed to interact with FMRFM In agreement with the recent work, we define biochemically that SMN forms droplets in vitro and these droplets are stabilized by RNA, suggesting that SMN could be involved in the formation of membraneless organelles, such as Cajal nuclear bodies. Finally, we found that SMN and FMRP co-fractionate with polysomes, in an RNA-dependent manner, suggesting a potential role in localized translation in motor neurons.


Subject(s)
Fragile X Mental Retardation Protein , Muscular Atrophy, Spinal , Survival of Motor Neuron 1 Protein , Humans , Infant , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Proteomics , RNA/metabolism , Survival of Motor Neuron 1 Protein/genetics
19.
Nat Commun ; 14(1): 7384, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37968267

ABSTRACT

Spinal muscular atrophy is an autosomal recessive neuromuscular disease caused by mutations in the multifunctional protein Survival of Motor Neuron, or SMN. Within the nucleus, SMN localizes to Cajal bodies, which are associated with nucleoli, nuclear organelles dedicated to the first steps of ribosome biogenesis. The highly organized structure of the nucleolus can be dynamically altered by genotoxic agents. RNAP1, Fibrillarin, and nucleolar DNA are exported to the periphery of the nucleolus after genotoxic stress and, once DNA repair is fully completed, the organization of the nucleolus is restored. We find that SMN is required for the restoration of the nucleolar structure after genotoxic stress. During DNA repair, SMN shuttles from the Cajal bodies to the nucleolus. This shuttling is important for nucleolar homeostasis and relies on the presence of Coilin and the activity of PRMT1.


Subject(s)
Muscular Atrophy, Spinal , RNA-Binding Proteins , Humans , RNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Cell Nucleolus/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Motor Neurons/metabolism , SMN Complex Proteins/metabolism , Coiled Bodies/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism
20.
J Neurosci ; 30(34): 11288-99, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20739549

ABSTRACT

Spinal muscular atrophy (SMA), a lethal neurodegenerative disease that occurs in childhood, is caused by the misexpression of the survival of motor neuron (SMN) protein in motor neurons. It is still unclear whether activating motor units in SMA corrects the delay in the postnatal maturation of the motor unit resulting in an enhanced neuroprotection. In the present work, we demonstrate that an adequate NMDA receptor activation in a type 2 SMA mouse model significantly accelerated motor unit postnatal maturation, counteracted apoptosis in the spinal cord, and induced a marked increase of SMN expression resulting from a modification of SMN2 gene transcription pattern. These beneficial effects were dependent on the level of NMDA receptor activation since a treatment with high doses of NMDA led to an acceleration of the motor unit maturation but favored the apoptotic process and decreased SMN expression. In addition, these results suggest that the NMDA-induced acceleration of motor unit postnatal maturation occurred independently of SMN. The NMDA receptor activating treatment strongly extended the life span in two different mouse models of severe SMA. The analysis of the intracellular signaling cascade that lay downstream the activated NMDA receptor revealed an unexpected reactivation of the CaMKII/AKT/CREB (cAMP response element-binding protein) pathway that induced an enhanced SMN expression. Therefore, pharmacological activation of spinal NMDA receptors could constitute a useful strategy for both increasing SMN expression and limiting motor neuron death in SMA spinal cord.


Subject(s)
Motor Neurons/physiology , Muscle Fibers, Skeletal/physiology , Muscular Atrophy, Spinal/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/growth & development , Survival of Motor Neuron 2 Protein/biosynthesis , Animals , Coculture Techniques , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Motor Neurons/drug effects , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/prevention & control , N-Methylaspartate/pharmacology , N-Methylaspartate/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate/agonists , Severity of Illness Index , Spinal Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL