Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 6408, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302774

ABSTRACT

Thyroid hormones (TH) regulate systemic glucose metabolism through incompletely understood mechanisms. Here, we show that improved glucose metabolism in hypothyroid mice after T3 treatment is accompanied with increased glucagon-like peptide-1 (GLP-1) production and insulin secretion, while co-treatment with a GLP-1 receptor antagonist attenuates the effects of T3 on insulin and glucose levels. By using mice lacking hepatic TH receptor ß (TRß) and a liver-specific TRß-selective agonist, we demonstrate that TRß-mediated hepatic TH signalling is required for both the regulation of GLP-1 production and the insulinotropic and glucose-lowering effects of T3. Moreover, administration of a liver-targeted TRß-selective agonist increases GLP-1 and insulin levels and alleviates hyperglycemia in diet-induced obesity. Mechanistically, T3 suppresses Cyp8b1 expression, resulting in increased the levels of Farnesoid X receptor (FXR)-antagonistic bile acids, thereby potentiating GLP-1 production and insulin secretion by repressing intestinal FXR signalling. T3 correlates with both plasma GLP-1 and fecal FXR-antagonistic bile acid levels in people with normal thyroid function. Thus, our study reveals a role for hepatic TH signalling in glucose homeostasis through the regulation of GLP-1 production via bile acid-mediated FXR antagonism.


Subject(s)
Bile Acids and Salts , Glucagon-Like Peptide 1 , Animals , Mice , Glucagon-Like Peptide 1/metabolism , Glucose , Homeostasis , Insulin , Liver/metabolism , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear , Receptors, G-Protein-Coupled/metabolism , Thyroid Hormones , Fragile X Mental Retardation Protein/antagonists & inhibitors
2.
Am J Physiol Cell Physiol ; 322(2): C125-C135, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34817267

ABSTRACT

Fragile X syndrome (FXS) is a genetic disorder that is characterized by a range of cognitive and behavioral deficits, including mild-moderate intellectual disability. The disease is characterized by an X-linked mutation of the Fmr1 gene, which causes silencing of the gene coding for fragile X mental retardation protein (FMRP), a translational regulator integral for neurodevelopment. Mitochondrial dysfunction has been recently associated with FXS, with reports of increases in oxidative stress markers, reactive oxygen species, and lipid peroxidation being present in the brain tissue. Astrocytes, a prominent glial cell within the central nervous system (CNS), play a large role in regulating oxidative homeostasis within the developing brain and dysregulation of astrocyte redox balance in FXS, which may contribute to oxidative stress. Astrocyte function and mitochondrial bioenergetics are significantly influenced by oxygen availability and circulating sex hormones; yet, these parameters are rarely considered during in vitro experimentation. Given that the brain normally develops in a range of hypoxic conditions and FXS is a sex-linked genetic disorder, we investigated how different oxygen levels (normoxic vs. hypoxic) and biological sex affected mitochondrial bioenergetics of astrocytes in FXS. Our results demonstrate that both mitochondrial respiration capacity and reactive oxygen species emission are altered with Fmr1 deletion in astrocytes and these changes were dependent upon both sexual dimorphism and oxygen availability.


Subject(s)
Astrocytes/metabolism , Energy Metabolism/physiology , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Mitochondria/metabolism , Sex Characteristics , Animals , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/metabolism , Female , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mitochondria/genetics , Reactive Oxygen Species/metabolism
3.
Nucleic Acids Res ; 49(16): 9479-9495, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34358321

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder caused by a limited expansion of CGG repeats in the FMR1 gene. Degeneration of neurons in FXTAS cell models can be triggered by accumulation of polyglycine protein (FMRpolyG), a by-product of translation initiated upstream to the repeats. Specific aims of our work included testing if naphthyridine-based molecules could (i) block FMRpolyG synthesis by binding to CGG repeats in RNA, (ii) reverse pathological alterations in affected cells and (iii) preserve the content of FMRP, translated from the same FMR1 mRNA. We demonstrate that cyclic mismatch binding ligand CMBL4c binds to RNA structure formed by CGG repeats and attenuates translation of FMRpolyG and formation of nuclear inclusions in cells transfected with vectors expressing RNA with expanded CGG repeats. Moreover, our results indicate that CMBL4c delivery can reduce FMRpolyG-mediated cytotoxicity and apoptosis. Importantly, its therapeutic potential is also observed once the inclusions are already formed. We also show that CMBL4c-driven FMRpolyG loss is accompanied by partial FMRP reduction. As complete loss of FMRP induces FXS in children, future experiments should aim at evaluation of CMBL4c therapeutic intervention in differentiated tissues, in which FMRpolyG translation inhibition might outweigh adverse effects related to FMRP depletion.


Subject(s)
Ataxia/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Naphthyridines/pharmacology , Tremor/genetics , Trinucleotide Repeat Expansion/drug effects , Apoptosis/drug effects , Ataxia/drug therapy , Ataxia/pathology , Cell Proliferation/drug effects , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Syndrome/drug therapy , Fragile X Syndrome/pathology , HeLa Cells , Humans , Ligands , Neurons/drug effects , Neurons/pathology , Peptides/genetics , Protein Biosynthesis/drug effects , Surface Plasmon Resonance , Tremor/drug therapy , Tremor/pathology , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/drug effects , Trinucleotide Repeats/genetics
4.
J Neurochem ; 158(5): 1094-1109, 2021 09.
Article in English | MEDLINE | ID: mdl-34327719

ABSTRACT

Fragile X mental retardation protein (FMRP) is a neuronal protein mediating multiple functions, with its absence resulting in one of the most common monogenic causes of autism, Fragile X syndrome (FXS). Analyses of FXS pathophysiology have identified a range of aberrations in synaptic signaling pathways and plasticity associated with group I metabotropic glutamate (mGlu) receptors. These studies, however, have mostly focused on the post-synaptic functions of FMRP and mGlu receptor activation, and relatively little is known about their presynaptic effects. Neurotransmitter release is mediated via multiple forms of synaptic vesicle (SV) fusion, each of which contributes to specific neuronal functions. The impacts of mGlu receptor activation and loss of FMRP on these SV fusion events remain unexplored. Here we combined electrophysiological and fluorescence imaging analyses on primary hippocampal cultures prepared from an Fmr1 knockout (KO) rat model. Compared to wild-type (WT) hippocampal neurons, KO neurons displayed an increase in the frequency of spontaneous excitatory post-synaptic currents (sEPSCs), as well as spontaneous SV fusion events. Pharmacological activation of mGlu receptors in WT neurons caused a similar increase in spontaneous SV fusion and sEPSC frequency. Notably, this increase in SV fusion was not observed when spontaneous activity was blocked using the sodium channel antagonist tetrodotoxin. Importantly, the effect of mGlu receptor activation on spontaneous SV fusion was occluded in Fmr1 KO neurons. Together, our results reveal that FMRP represses spontaneous presynaptic SV fusion, whereas mGlu receptor activation increases this event. This reciprocal control appears to be mediated via their regulation of intrinsic neuronal excitability.


Subject(s)
Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Fragile X Mental Retardation Protein/genetics , Male , Membrane Fusion/physiology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, Metabotropic Glutamate/genetics , Synaptic Vesicles/genetics
5.
Neurobiol Dis ; 154: 105338, 2021 07.
Article in English | MEDLINE | ID: mdl-33775821

ABSTRACT

Fragile X syndrome (FXS) is a common form of intellectual disability and autism caused by the lack of Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein involved in RNA transport and protein synthesis. Upon cellular stress, global protein synthesis is blocked and mRNAs are recruited into stress granules (SGs), together with RNA-binding proteins including FMRP. Activation of group-I metabotropic glutamate (mGlu) receptors stimulates FMRP-mediated mRNA transport and protein synthesis, but their role in SGs formation is unexplored. To this aim, we pre-treated wild type (WT) and Fmr1 knockout (KO) cultured astrocytes with the group-I-mGlu receptor agonist (S)-3,5-Dihydroxyphenylglycine (DHPG) and exposed them to sodium arsenite (NaAsO2), a widely used inducer of SGs formation. In WT cultures the activation of group-I mGlu receptors reduced SGs formation and recruitment of FMRP into SGs, and also attenuated phosphorylation of eIF2α, a key event crucially involved in SGs formation and inhibition of protein synthesis. In contrast, Fmr1 KO astrocytes, which exhibited a lower number of SGs than WT astrocytes, did not respond to agonist stimulation. Interestingly, the mGlu5 receptor negative allosteric modulator (NAM) 2-methyl-6-(phenylethynyl)pyridine (MPEP) antagonized DHPG-mediated SGs reduction in WT and reversed SGs formation in Fmr1 KO cultures. Our findings reveal a novel function of mGlu5 receptor as modulator of SGs formation and open new perspectives for understanding cellular response to stress in FXS pathophysiology.


Subject(s)
Astrocytes/metabolism , Fragile X Mental Retardation Protein/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Stress Granules/metabolism , Animals , Animals, Newborn , Astrocytes/pathology , Cells, Cultured , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Mice , Mice, Knockout , Oxidative Stress/physiology , Receptor, Metabotropic Glutamate 5/genetics , Stress Granules/pathology
6.
Cell Mol Gastroenterol Hepatol ; 11(2): 639-658, 2021.
Article in English | MEDLINE | ID: mdl-33091622

ABSTRACT

BACKGROUND & AIMS: The fragile X mental retardation protein (FMRP) affects multiple steps of the mRNA metabolism during brain development and in different neoplastic processes. However, the contribution of FMRP in colon carcinogenesis has not been investigated. METHODS: FMR1 mRNA transcript and FMRP protein expression were analyzed in human colon samples derived from patients with sporadic colorectal cancer (CRC) and healthy subjects. We used a well-established mouse model of sporadic CRC induced by azoxymethane to determine the possible role of FMRP in CRC. To address whether FMRP controls cancer cell survival, we analyzed cell death pathway in CRC human epithelial cell lines and in patient-derived colon cancer organoids in presence or absence of a specific FMR1 antisense oligonucleotide or siRNA. RESULTS: We document a significant increase of FMRP in human CRC relative to non-tumor tissues. Next, using an inducible mouse model of CRC, we observed a reduction of colonic tumor incidence and size in the Fmr1 knockout mice. The abrogation of FMRP induced spontaneous cell death in human CRC cell lines activating the necroptotic pathway. Indeed, specific immunoprecipitation experiments on human cell lines and CRC samples indicated that FMRP binds receptor-interacting protein kinase 1 (RIPK1) mRNA, suggesting that FMRP acts as a regulator of necroptosis pathway through the surveillance of RIPK1 mRNA metabolism. Treatment of human CRC cell lines and patient-derived colon cancer organoids with the FMR1 antisense resulted in up-regulation of RIPK1. CONCLUSIONS: Altogether, these data support a role for FMRP  in controlling RIPK1 expression and necroptotic activation in CRC.


Subject(s)
Colorectal Neoplasms/genetics , Fragile X Mental Retardation Protein/metabolism , Neoplasm Recurrence, Local/epidemiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Azoxymethane/administration & dosage , Azoxymethane/toxicity , Carcinogenesis/genetics , Case-Control Studies , Cell Culture Techniques , Cell Line, Tumor , Colon/pathology , Colon/surgery , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Datasets as Topic , Disease-Free Survival , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Healthy Volunteers , Humans , Male , Mice , Mice, Knockout , Necroptosis/genetics , Neoplasm Recurrence, Local/genetics , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Organoids , Prognosis
7.
Genes (Basel) ; 11(4)2020 03 27.
Article in English | MEDLINE | ID: mdl-32230785

ABSTRACT

In fragile X syndrome (FXS), expansion of a CGG repeat tract in the 5'-untranslated region of the FMR1 gene to >200 repeats causes transcriptional silencing by inducing heterochromatin formation. Understanding the mechanism of FMR1 silencing is important as gene reactivation is a potential treatment approach for FXS. To date, only the DNA demethylating drug 5-azadeoxycytidine (AZA) has proved effective at gene reactivation; however, this drug is toxic. The repressive H3K9 methylation mark is enriched on the FMR1 gene in FXS patient cells and is thus a potential druggable target. However, its contribution to the silencing process is unclear. Here, we studied the effect of small molecule inhibitors of H3K9 methylation on FMR1 expression in FXS patient cells. Chaetocin showed a small effect on FMR1 gene reactivation and a synergistic effect on FMR1 mRNA levels when used in combination with AZA. Additionally, chaetocin, BIX01294 and 3-Deazaneplanocin A (DZNep) were able to significantly delay the re-silencing of AZA-reactivated FMR1 alleles. These data are consistent with the idea that H3K9 methylation precedes DNA methylation and that removal of DNA methylation is necessary to see the optimal effect of histone methyl-transferase (HMT) inhibitors on FMR1 gene expression. Nonetheless, our data also show that drugs targeting repressive H3K9 methylation marks are able to produce sustained reactivation of the FMR1 gene after a single dose of AZA.


Subject(s)
DNA Methylation , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Syndrome/genetics , Gene Silencing , Pharmaceutical Preparations/metabolism , Small Molecule Libraries/pharmacology , Alleles , Cells, Cultured , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/drug therapy , Fragile X Syndrome/pathology , Humans , Trinucleotide Repeats
8.
J Biol Chem ; 294(52): 19889-19895, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31753916

ABSTRACT

N6-Methyladenosine (m6A) is the most abundant post-transcriptional mRNA modification in eukaryotes and exerts many of its effects on gene expression through reader proteins that bind specifically to m6A-containing transcripts. Fragile X mental retardation protein (FMRP), an RNA-binding protein, has previously been shown to affect the translation of target mRNAs and trafficking of mRNA granules. Loss of function of FMRP causes fragile X syndrome, the most common form of inherited intellectual disability in humans. Using HEK293T cells, siRNA-mediated gene knockdown, cytoplasmic and nuclear fractions, RNA-Seq, and LC-MS/MS analyses, we demonstrate here that FMRP binds directly to a collection of m6A sites on mRNAs. FMRP depletion increased mRNA m6A levels in the nucleus. Moreover, the abundance of FMRP targets in the cytoplasm relative to the nucleus was decreased in Fmr1-KO mice, an effect also observed in highly methylated genes. We conclude that FMRP may affect the nuclear export of m6A-modified RNA targets.


Subject(s)
Adenosine/analogs & derivatives , Fragile X Mental Retardation Protein/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Adenosine/metabolism , Animals , Binding Sites , Cell Nucleus/metabolism , Cerebral Cortex/metabolism , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA Stability , RNA, Messenger/chemistry , RNA, Small Interfering/metabolism
9.
J Neurosci ; 39(23): 4595-4605, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30940719

ABSTRACT

An essential aspect of goal-directed decision-making is selecting actions based on anticipated consequences, a process that involves the orbitofrontal cortex (OFC) and potentially, the plasticity of dendritic spines in this region. To investigate this possibility, we trained male and female mice to nose poke for food reinforcers, or we delivered the same number of food reinforcers non-contingently to separate mice. We then decreased the likelihood of reinforcement for trained mice, requiring them to modify action-outcome expectations. In a separate experiment, we blocked action-outcome updating via chemogenetic inactivation of the OFC. In both cases, successfully selecting actions based on their likely consequences was associated with fewer immature, thin-shaped dendritic spines and a greater proportion of mature, mushroom-shaped spines in the ventrolateral OFC. This pattern was distinct from spine loss associated with aging, and we identified no effects on hippocampal CA1 neurons. Given that the OFC is involved in prospective calculations of likely outcomes, even when they are not observable, constraining spinogenesis while preserving mature spines may be important for solidifying durable expectations. To investigate causal relationships, we inhibited the RNA-binding protein fragile X mental retardation protein (encoded by Fmr1), which constrains dendritic spine turnover. Ventrolateral OFC-selective Fmr1 knockdown recapitulated the behavioral effects of inducible OFC inactivation (and lesions; also shown here), impairing action-outcome conditioning, and caused dendritic spine excess. Our findings suggest that a proper balance of dendritic spine plasticity within the OFC is necessary for one's ability to select actions based on anticipated consequences.SIGNIFICANCE STATEMENT Navigating a changing environment requires associating actions with their likely outcomes and updating these associations when they change. Dendritic spine plasticity is likely involved, yet relationships are unconfirmed. Using behavioral, chemogenetic, and viral-mediated gene silencing strategies and high-resolution microscopy, we find that modifying action-outcome expectations is associated with fewer immature spines and a greater proportion of mature spines in the ventrolateral orbitofrontal cortex (OFC). Given that the OFC is involved in prospectively calculating the likely outcomes of one's behavior, even when they are not observable, constraining spinogenesis while preserving mature spines may be important for maintaining durable expectations.


Subject(s)
Anticipation, Psychological/physiology , Dendritic Spines/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Reward , Animals , Conditioning, Operant , Decision Making , Dendritic Spines/ultrastructure , Dependovirus/genetics , Feeding Behavior , Female , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/physiology , Gene Knockdown Techniques , Genes, Reporter , Genetic Vectors/administration & dosage , Male , Mice , Mice, Inbred C57BL , Optogenetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Reinforcement, Psychology
10.
Cell Rep ; 16(9): 2281-8, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545877

ABSTRACT

Human mutations in the cytoplasmic C-terminal domain of Slack sodium-activated potassium (KNa) channels result in childhood epilepsy with severe intellectual disability. Slack currents can be increased by pharmacological activators or by phosphorylation of a Slack C-terminal residue by protein kinase C. Using an optical biosensor assay, we find that Slack channel stimulation in neurons or transfected cells produces loss of mass near the plasma membrane. Slack mutants associated with intellectual disability fail to trigger any change in mass. The loss of mass results from the dissociation of the protein phosphatase 1 (PP1) targeting protein, Phactr-1, from the channel. Phactr1 dissociation is specific to wild-type Slack channels and is not observed when related potassium channels are stimulated. Our findings suggest that Slack channels are coupled to cytoplasmic signaling pathways and that dysregulation of this coupling may trigger the aberrant intellectual development associated with specific childhood epilepsies.


Subject(s)
Cell Membrane/metabolism , Fragile X Mental Retardation Protein/genetics , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Potassium Channels/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biosensing Techniques , Bithionol/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Membrane/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Ion Transport/drug effects , Mice , Mice, Knockout , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/metabolism , Mutation , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Phosphorylation , Potassium Channels/agonists , Potassium Channels/metabolism , Potassium Channels, Sodium-Activated , Primary Cell Culture , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Thiazolidines/pharmacology , Xenopus laevis
11.
Biofactors ; 41(2): 121-6, 2015.
Article in English | MEDLINE | ID: mdl-25809670

ABSTRACT

Cyclic adenosine monophosphate (cAMP) modulates synaptic plasticity and memory and manipulation of the cAMP/protein kinase A/cAMP responsive element binding protein pathway significantly affects cognitive functions. Notably, cAMP can increase the expression of the amyloid precursor protein (APP), whose proteolytic processing gives rise to amyloid beta (Aß) peptides. Despite playing a pathogenic role in Alzheimer's disease, physiological concentrations of Aß are necessary for the cAMP-mediated regulation of long-term potentiation, supporting the existence of a novel cAMP/APP/Aß cascade with a crucial role in memory formation. However, the molecular mechanisms by which cAMP stimulates APP expression and Aß production remain unclear. Here, we investigated whether hnRNP-C and FMRP, two RNA-binding proteins largely involved in the expression of APP, are the cAMP effectors inducing the protein synthesis of APP. Using RNA immunoprecipitation and RNA-silencing approaches, we found that neither hnRNP-C nor FMRP is required for cAMP to stimulate APP and Aß production.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Cyclic AMP/metabolism , Fragile X Mental Retardation Protein/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Neurons/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Animals , Cell Line , Colforsin/pharmacology , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein Group C/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Humans , Mice , Neurons/cytology , Neurons/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
12.
Hum Mol Genet ; 23(24): 6575-83, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25055869

ABSTRACT

The FMR1 gene is subject to repeat mediated-gene silencing when the CGG-repeat tract in the 5' UTR exceeds 200 repeat units. This results in Fragile X syndrome, the most common heritable cause of intellectual disability and a major cause of autism spectrum disorders. The mechanism of gene silencing is not fully understood, and efforts to reverse this gene silencing have had limited success. Here, we show that the level of trimethylation of histone H3 on lysine 27, a hallmark of the activity of EZH2, a component of repressive Polycomb Group (PcG) complexes like PRC2, is increased on reactivation of the silenced allele by either the DNA demethylating agent 5-azadeoxycytidine or the SIRT1 inhibitor splitomicin. The level of H3K27me3 increases and decreases in parallel with the FMR1 mRNA level. Furthermore, reducing the levels of the FMR1 mRNA reduces the accumulation of H3K27me3. This suggests a model for FMR1 gene silencing in which the FMR1 mRNA generated from the reactivated allele acts in cis to repress its own transcription via the recruitment of PcG complexes to the FMR1 locus.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Histones/genetics , Lymphocytes/metabolism , Polycomb Repressive Complex 2/genetics , RNA, Messenger/genetics , Transcription, Genetic , Alleles , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line , Decitabine , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/pharmacology , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Gene Expression Regulation , Histones/metabolism , Humans , Lymphocytes/drug effects , Lymphocytes/pathology , Male , Naphthalenes/pharmacology , Polycomb Repressive Complex 2/metabolism , Pyrones/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/metabolism
13.
EMBO Mol Med ; 5(10): 1523-36, 2013 10.
Article in English | MEDLINE | ID: mdl-24092663

ABSTRACT

The role of the fragile X mental retardation protein (FMRP) is well established in brain, where its absence leads to the fragile X syndrome (FXS). FMRP is almost ubiquitously expressed, suggesting that, in addition to its effects in brain, it may have fundamental roles in other organs. There is evidence that FMRP expression can be linked to cancer. FMR1 mRNA, encoding FMRP, is overexpressed in hepatocellular carcinoma cells. A decreased risk of cancer has been reported in patients with FXS while a patient-case with FXS showed an unusual decrease of tumour brain invasiveness. However, a role for FMRP in regulating cancer biology, if any, remains unknown. We show here that FMRP and FMR1 mRNA levels correlate with prognostic indicators of aggressive breast cancer, lung metastases probability and triple negative breast cancer (TNBC). We establish that FMRP overexpression in murine breast primary tumours enhances lung metastasis while its reduction has the opposite effect regulating cell spreading and invasion. FMRP binds mRNAs involved in epithelial mesenchymal transition (EMT) and invasion including E-cadherin and Vimentin mRNAs, hallmarks of EMT and cancer progression.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , RNA, Messenger/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Shape , Disease Progression , Epithelial-Mesenchymal Transition , Female , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , RNA Interference , RNA, Small Interfering/metabolism , Vimentin/metabolism
14.
Cell Death Dis ; 4: e617, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23640459

ABSTRACT

Our previous studies have shown that microRNA-383 (miR-383) expression is downregulated in the testes of infertile men with maturation arrest (MA). Abnormal testicular miR-383 expression may potentiate the connections between male infertility and testicular germ cell tumors. However, the mechanisms underlying the targeting and functions of miR-383 during spermatogenesis remain unknown. In this study, we found that fragile X mental retardation protein (FMRP) was associated with 88 miRNAs in mouse testis including miR-383. Knockdown of FMRP in NTERA-2 (NT2) (testicular embryonal carcinoma) cells enhanced miR-383-induced suppression of cell proliferation by decreasing the interaction between FMRP and miR-383, and then affecting miR-383 binding to the 3'-untranslated region of its target genes, including interferon regulatory factor-1 (IRF1) and Cyclin D1 both in vivo and in vitro. On the other hand, FMRP levels were also downregulated by overexpression of miR-383 in NT2 cells and GC1 (spermatogonia germ cell line). miR-383 targeted to Cyclin D1 directly, and then inhibited its downstream effectors, including phosphorylated pRb and E2F1, which ultimately resulted in decreased FMRP expression. Reduced miR-383 expression, dysregulated cyclin-dependent kinase 4 expression (one of the downstream genes of miR-383) and increased DNA damage were also observed in the testes of Fmr1 knockout mice and of MA patients with a downregulation of FMRP. A potential feedback loop between FMRP and miR-383 during spermatogenesis is proposed, and FMRP acts as a negative regulator of miR-383 functions. Our data also indicate that dysregulation of the FMRP-miR-383 pathway may partially contribute to human spermatogenic failure with MA.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , MicroRNAs/metabolism , Spermatogenesis/genetics , 3' Untranslated Regions , Animals , Base Sequence , Cell Line , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Down-Regulation , E2F1 Transcription Factor/metabolism , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Male , Mice , Mice, Knockout , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Testis/metabolism
15.
J Neurosci ; 27(20): 5338-48, 2007 May 16.
Article in English | MEDLINE | ID: mdl-17507556

ABSTRACT

Fragile X syndrome, a common form of inherited mental retardation, is caused by the loss of fragile X mental retardation protein (FMRP), an mRNA binding protein that is hypothesized to regulate local mRNA translation in dendrites downstream of gp1 metabotropic glutamate receptors (mGluRs). However, specific FMRP-associated mRNAs that localize to dendrites in vivo and show altered mGluR-dependent translation at synapses of Fmr1 knock-out mice are unknown so far. Using fluorescence in situ hybridization, we discovered that GluR1/2 and postsynaptic density-95 (PSD-95) mRNAs are localized to dendrites of cortical and hippocampal neurons in vivo. Quantitative analyses of their dendritic mRNA levels in cultured neurons and synaptoneurosomes did not detect differences between wild-type and Fmr1 knock-out (KO) mice. In contrast, PSD-95, GluR1/2, and calcium/calmodulin-dependent kinase IIalpha (CaMKIIalpha) mRNA levels in actively translating polyribosomes were dysregulated in synaptoneurosomes from Fmr1 knock-out mice in response to mGluR activation. [35S]methionine incorporation into newly synthesized proteins similarly revealed impaired stimulus-induced protein synthesis of CaMKIIalpha and PSD-95 in synaptoneurosomes from Fmr1 KO mice. Quantitative analysis of mRNA levels in FMRP-specific immunoprecipitations from synaptoneurosomes demonstrated the association of FMRP with CaMKIIalpha, PSD-95, and GluR1/2 mRNAs. These findings suggest a novel mechanism whereby FMRP regulates the local synthesis AMPA receptor (AMPAR) subunits, PSD-95, and CaMKIIalpha downstream of mGluR-activation. Dysregulation of local translation of AMPAR and associated factors at synapses may impair control of the molecular composition of the postsynaptic density and consequently alter synaptic transmission, causing impairments of neuronal plasticity observed in Fmr1 knock-out mice and fragile X syndrome.


Subject(s)
Fragile X Syndrome/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/biosynthesis , Animals , Disease Models, Animal , Disks Large Homolog 4 Protein , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Guanylate Kinases , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, AMPA/genetics , Synapses/genetics , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL