Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.623
Filter
1.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230221, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853554

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and is the leading known single-gene cause of autism spectrum disorder. Patients with FXS display varied behavioural deficits that include mild to severe cognitive impairments in addition to mood disorders. Currently, there is no cure for this condition; however, there is an emerging focus on therapies that inhibit mechanistic target of rapamycin (mTOR)-dependent protein synthesis owing to the clinical effectiveness of metformin for alleviating some behavioural symptoms in FXS. Adiponectin (APN) is a neurohormone that is released by adipocytes and provides an alternative means to inhibit mTOR activation in the brain. In these studies, we show that Fmr1 knockout mice, like patients with FXS, show reduced levels of circulating APN and that both long-term potentiation (LTP) and long-term depression (LTD) in the dentate gyrus (DG) are impaired. Brief (20 min) incubation of hippocampal slices in APN (50 nM) was able to rescue both LTP and LTD in the DG and increased both the surface expression and phosphorylation of GluA1 receptors. These results provide evidence for reduced APN levels in FXS playing a role in decreasing bidirectional synaptic plasticity and show that therapies which enhance APN levels may have therapeutic potential for this and related conditions.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Adiponectin , Dentate Gyrus , Disease Models, Animal , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neuronal Plasticity , Animals , Fragile X Syndrome/physiopathology , Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/drug effects , Mice , Neuronal Plasticity/drug effects , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Adiponectin/metabolism , Long-Term Potentiation/drug effects , Male , Receptors, AMPA/metabolism
2.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230484, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853552

ABSTRACT

Fragile X syndrome (FXS) is characterized by impairments in executive function including different types of learning and memory. Long-term potentiation (LTP), thought to underlie the formation of memories, has been studied in the Fmr1 mouse model of FXS. However, there have been many discrepancies in the literature with inconsistent use of littermate and non-littermate Fmr1 knockout (KO) and wild-type (WT) control mice. Here, the influence of the breeding strategy (cage effect) on short-term potentiation (STP), LTP, contextual fear conditioning (CFC), expression of N-methyl-d-aspartate receptor (NMDAR) subunits and the modulation of NMDARs, were examined. The largest deficits in STP, LTP and CFC were found in KO mice compared with non-littermate WT. However, the expression of NMDAR subunits was unchanged in this comparison. Rather, NMDAR subunit (GluN1, 2A, 2B) expression was sensitive to the cage effect, with decreased expression in both WT and KO littermates compared with non-littermates. Interestingly, an NMDAR-positive allosteric modulator, UBP714, was only effective in potentiating the induction of LTP in non-littermate KO mice and not the littermate KO mice. These results suggest that commonly studied phenotypes in Fmr1 KOs are sensitive to the cage effect and therefore the breeding strategy may contribute to discrepancies in the literature.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Disease Models, Animal , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate , Animals , Fragile X Syndrome/physiopathology , Fragile X Syndrome/genetics , Mice , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Long-Term Potentiation , Male , Mice, Inbred C57BL , Housing, Animal , Fear
3.
J Ovarian Res ; 17(1): 103, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760837

ABSTRACT

BACKGROUND: Fragile X-associated primary ovarian insufficiency (FXPOI), characterized by amenorrhea before age 40 years, occurs in 20% of female FMR1 premutation carriers. Presently, there are no molecular or biomarkers that can help predicting which FMR1 premutation women will develop FXPOI. We previously demonstrated that high FMR4 levels can discriminate between FMR1 premutation carriers with and without FXPOI. In the present study the relationship between the expression levels of FMR4 and the ovarian reserve markers was assessed in female FMR1 premutation carriers under age of 35 years. METHODS: We examined the association between FMR4 transcript levels and the measures of total antral follicle count (AFC) and serum anti-müllerian hormone (AMH) levels as markers of ovarian follicle reserve. RESULTS: Results revealed a negative association between FMR4 levels and AMH (r = 0.45) and AFC (r = 0.64). Statistically significant higher FMR4 transcript levels were found among those FMR1 premutation women with both, low AFCs and AMH levels. CONCLUSIONS: These findings reinforce previous studies supporting the association between high levels of FMR4 and the risk of developing FXPOI in FMR1 premutation carriers.


Subject(s)
Anti-Mullerian Hormone , Biomarkers , Fragile X Mental Retardation Protein , Ovarian Reserve , Primary Ovarian Insufficiency , Humans , Female , Fragile X Mental Retardation Protein/genetics , Ovarian Reserve/genetics , Adult , Biomarkers/blood , Anti-Mullerian Hormone/blood , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/blood , Heterozygote , Fragile X Syndrome/genetics , Fragile X Syndrome/blood , Mutation , Ovarian Follicle/metabolism , Young Adult
4.
BMC Neurol ; 24(1): 154, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714961

ABSTRACT

BACKGROUND: Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder caused by CGG repeat expansion of FMR1 gene. Both FXTAS and neuronal intranuclear inclusion disease (NIID) belong to polyglycine diseases and present similar clinical, radiological, and pathological features, making it difficult to distinguish these diseases. Reversible encephalitis-like attacks are often observed in NIID. It is unclear whether they are presented in FXTAS and can be used for differential diagnosis of NIID and FXTAS. CASE PRESENTATION: A 63-year-old Chinese male with late-onset gait disturbance, cognitive decline, and reversible attacks of fever, consciousness impairment, dizziness, vomiting, and urinary incontinence underwent neurological assessment and examinations, including laboratory tests, electroencephalogram test, imaging, skin biopsy, and genetic test. Brain MRI showed T2 hyperintensities in middle cerebellar peduncle and cerebrum, in addition to cerebellar atrophy and DWI hyperintensities along the corticomedullary junction. Lesions in the brainstem were observed. Skin biopsy showed p62-positive intranuclear inclusions. The possibilities of hypoglycemia, lactic acidosis, epileptic seizures, and cerebrovascular attacks were excluded. Genetic analysis revealed CGG repeat expansion in FMR1 gene, and the number of repeats was 111. The patient was finally diagnosed as FXTAS. He received supportive treatment as well as symptomatic treatment during hospitalization. His encephalitic symptoms were completely relieved within one week. CONCLUSIONS: This is a detailed report of a case of FXTAS with reversible encephalitis-like episodes. This report provides new information for the possible and rare features of FXTAS, highlighting that encephalitis-like episodes are common in polyglycine diseases and unable to be used for differential diagnosis.


Subject(s)
Ataxia , Encephalitis , Fragile X Syndrome , Tremor , Humans , Male , Middle Aged , Tremor/diagnosis , Tremor/genetics , Tremor/etiology , Fragile X Syndrome/genetics , Fragile X Syndrome/diagnosis , Fragile X Syndrome/complications , Ataxia/diagnosis , Ataxia/genetics , Encephalitis/diagnosis , Encephalitis/complications , Encephalitis/genetics , Encephalitis/pathology , Fragile X Mental Retardation Protein/genetics , Diagnosis, Differential , Intranuclear Inclusion Bodies/pathology , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/complications
5.
Cereb Cortex ; 34(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38771240

ABSTRACT

In vitro and ex vivo studies have shown consistent indications of hyperexcitability in the Fragile X Messenger Ribonucleoprotein 1 (Fmr1) knockout mouse model of autism spectrum disorder. We recently introduced a method to quantify network-level functional excitation-inhibition ratio from the neuronal oscillations. Here, we used this measure to study whether the implicated synaptic excitation-inhibition disturbances translate to disturbances in network physiology in the Fragile X Messenger Ribonucleoprotein 1 (Fmr1) gene knockout model. Vigilance-state scoring was used to extract segments of inactive wakefulness as an equivalent behavioral condition to the human resting-state and, subsequently, we performed high-frequency resolution analysis of the functional excitation-inhibition biomarker, long-range temporal correlations, and spectral power. We corroborated earlier studies showing increased high-frequency power in Fragile X Messenger Ribonucleoprotein 1 (Fmr1) knockout mice. Long-range temporal correlations were higher in the gamma frequency ranges. Contrary to expectations, functional excitation-inhibition was lower in the knockout mice in high frequency ranges, suggesting more inhibition-dominated networks. Exposure to the Gamma-aminobutyric acid (GABA)-agonist clonazepam decreased the functional excitation-inhibition in both genotypes, confirming that increasing inhibitory tone results in a reduction of functional excitation-inhibition. In addition, clonazepam decreased electroencephalogram power and increased long-range temporal correlations in both genotypes. These findings show applicability of these new resting-state electroencephalogram biomarkers to animal for translational studies and allow investigation of the effects of lower-level disturbances in excitation-inhibition balance.


Subject(s)
Fragile X Mental Retardation Protein , Mice, Knockout , Neurons , Animals , Fragile X Mental Retardation Protein/genetics , Neurons/physiology , Neurons/drug effects , Neurons/metabolism , Mice , Male , Neural Inhibition/physiology , Neural Inhibition/drug effects , Mice, Inbred C57BL , Electroencephalography
6.
Dev Neuropsychol ; 49(4): 153-166, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38753030

ABSTRACT

Although most individuals who carry the Fragile X premutation allele, defined as 55-200 CGG repeats on the X-linked FMR1 gene (Fragile X Messenger Ribonucleoprotein 1 gene), do not meet diagnostic criteria for autism spectrum disorder, there is a suggestion of increased behaviors associated with subtle autistic traits. More autism associated characteristics have been reported among adults than children. This may highlight a possible worsening developmental trajectory, variable findings due to research quality or differences in number of studies done in adults vs children, rather than true developmental changes. This review is designed to examine the neurodevelopmental profile associated with the premutation allele from a developmental perspective, focused on autistic traits.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Humans , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Child , Alleles , Autism Spectrum Disorder/genetics , Adult , Autistic Disorder/genetics
7.
Elife ; 122024 May 17.
Article in English | MEDLINE | ID: mdl-38757694

ABSTRACT

The fragile X syndrome (FXS) represents the most prevalent form of inherited intellectual disability and is the first monogenic cause of autism spectrum disorder. FXS results from the absence of the RNA-binding protein FMRP (fragile X messenger ribonucleoprotein). Neuronal migration is an essential step of brain development allowing displacement of neurons from their germinal niches to their final integration site. The precise role of FMRP in neuronal migration remains largely unexplored. Using live imaging of postnatal rostral migratory stream (RMS) neurons in Fmr1-null mice, we observed that the absence of FMRP leads to delayed neuronal migration and altered trajectory, associated with defects of centrosomal movement. RNA-interference-induced knockdown of Fmr1 shows that these migratory defects are cell-autonomous. Notably, the primary Fmrp mRNA target implicated in these migratory defects is microtubule-associated protein 1B (MAP1B). Knocking down MAP1B expression effectively rescued most of the observed migratory defects. Finally, we elucidate the molecular mechanisms at play by demonstrating that the absence of FMRP induces defects in the cage of microtubules surrounding the nucleus of migrating neurons, which is rescued by MAP1B knockdown. Our findings reveal a novel neurodevelopmental role for FMRP in collaboration with MAP1B, jointly orchestrating neuronal migration by influencing the microtubular cytoskeleton.


Subject(s)
Cell Movement , Fragile X Mental Retardation Protein , Mice, Knockout , Microtubule-Associated Proteins , Neurons , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Animals , Neurons/metabolism , Neurons/physiology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mice , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Gene Knockdown Techniques
8.
Psychiatry Res ; 337: 115962, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763080

ABSTRACT

Fragile X Syndrome (FXS) results from the silencing of the FMR1 gene and is the most prevalent inherited cause of intellectual disability and the most frequent monogenic cause of autism spectrum disorder. It is well established that Fragile X individuals are subjected to a wide array of comorbidities, ranging from cognitive, behavioural, and medical origin. Furthermore, recent studies have also described metabolic impairments in FXS individuals. However, the molecular mechanisms linking FMRP deficiency to improper metabolism are still misunderstood. The endocannabinoidome (eCBome) is a lipid-based signalling system that regulates several functions across the body, ranging from cognition, behaviour and metabolism. Alterations in the eCBome have been described in FXS animal models and linked to neuronal hyperexcitability, a core deficit of the disease. However, the potential link between dysregulation of the eCBome and altered metabolism observed in FXS remains unexplored. As such, this review aims to overcome this issue by describing the most recent finding related to eCBome and metabolic dysfunctions in the context of FXS. A better comprehension of this association will help deepen our understanding of FXS pathophysiology and pave the way for future therapeutic interventions.


Subject(s)
Endocannabinoids , Fragile X Syndrome , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Humans , Endocannabinoids/metabolism , Animals , Metabolic Networks and Pathways , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism
9.
Neurosci Biobehav Rev ; 162: 105731, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763180

ABSTRACT

Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Neuroglia , Humans , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Neuroglia/metabolism , Animals , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Fragile X Syndrome/pathology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Brain Diseases/genetics , Ataxia/metabolism , Ataxia/physiopathology , Ataxia/genetics , Tremor/metabolism , Tremor/physiopathology , Tremor/genetics
10.
J Neurodev Disord ; 16(1): 24, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720271

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is currently diagnosed in approximately 1 in 44 children in the United States, based on a wide array of symptoms, including sensory dysfunction and abnormal language development. Boys are diagnosed ~ 3.8 times more frequently than girls. Auditory temporal processing is crucial for speech recognition and language development. Abnormal development of temporal processing may account for ASD language impairments. Sex differences in the development of temporal processing may underlie the differences in language outcomes in male and female children with ASD. To understand mechanisms of potential sex differences in temporal processing requires a preclinical model. However, there are no studies that have addressed sex differences in temporal processing across development in any animal model of ASD. METHODS: To fill this major gap, we compared the development of auditory temporal processing in male and female wildtype (WT) and Fmr1 knock-out (KO) mice, a model of Fragile X Syndrome (FXS), a leading genetic cause of ASD-associated behaviors. Using epidural screw electrodes, we recorded auditory event related potentials (ERP) and auditory temporal processing with a gap-in-noise auditory steady state response (ASSR) paradigm at young (postnatal (p)21 and p30) and adult (p60) ages from both auditory and frontal cortices of awake, freely moving mice. RESULTS: The results show that ERP amplitudes were enhanced in both sexes of Fmr1 KO mice across development compared to WT counterparts, with greater enhancement in adult female than adult male KO mice. Gap-ASSR deficits were seen in the frontal, but not auditory, cortex in early development (p21) in female KO mice. Unlike male KO mice, female KO mice show WT-like temporal processing at p30. There were no temporal processing deficits in the adult mice of both sexes. CONCLUSIONS: These results show a sex difference in the developmental trajectories of temporal processing and hypersensitive responses in Fmr1 KO mice. Male KO mice show slower maturation of temporal processing than females. Female KO mice show stronger hypersensitive responses than males later in development. The differences in maturation rates of temporal processing and hypersensitive responses during various critical periods of development may lead to sex differences in language function, arousal and anxiety in FXS.


Subject(s)
Disease Models, Animal , Evoked Potentials, Auditory , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Sex Characteristics , Animals , Fragile X Syndrome/physiopathology , Female , Male , Mice , Evoked Potentials, Auditory/physiology , Fragile X Mental Retardation Protein/genetics , Auditory Perception/physiology , Autism Spectrum Disorder/physiopathology , Auditory Cortex/physiopathology , Mice, Inbred C57BL
11.
BMJ Case Rep ; 17(5)2024 May 27.
Article in English | MEDLINE | ID: mdl-38802254

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a progressive hereditary neurodegenerative disorder which causes intention tremor and cerebellar ataxia. It typically affects the ageing population. Deep brain stimulation (DBS) is widely accepted in the treatment of common movement disorders and has been trialled in treating rare and complex neurodegenerative disorders. We report a case of a man in his 40s with a long history of tremor affecting his hands. MRI brain revealed high T2 signal in the middle cerebellar peduncles. Genetic testing revealed FMR1 premutation confirming the diagnosis of FXTAS. Subsequently, he was treated with multitarget DBS of the ventralis intermediate nucleus and ventralis oralis posterior nuclei bilaterally, with excellent neurological function at 9 years follow-up. This case suggests multitarget DBS for FXTAS with neurophysiology-guided DBS programming can provide excellent long-term tremor suppression in selected patients.


Subject(s)
Ataxia , Deep Brain Stimulation , Fragile X Syndrome , Tremor , Humans , Male , Ataxia/therapy , Deep Brain Stimulation/methods , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/therapy , Magnetic Resonance Imaging , Tremor/therapy
12.
Mol Genet Genomic Med ; 12(4): e2425, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38562051

ABSTRACT

BACKGROUND: To explore the clinical application value of pre-conception expanded carrier screening (PECS) in the Chinese Han ethnicity population of childbearing age. METHODS: The results of genetic testing of infertile parents who underwent PECS in the Reproductive Medicine Center of the Second Affiliated Hospital of Zhengzhou University, China, from September 2019 to December 2021, were retrospectively analyzed. The carrier rate of single gene disease, the detection rate of high-risk parents, and the clinical outcome of high-risk parents were statistically analyzed. RESULTS: A total of 1372 Chinese Han ethnicity patients underwent PECS, among which 458 patients underwent the extended 108-gene test, their overall carrier rate was 31.7%, and the detection rate of high-risk parents was 0.3%. The highest carrier rates were SLC22A (2.4%), ATP7B (2.4%), MMACHC (2.2%), PAH (1.8%), GALC (1.8%), MLC1 (1.3%), UNC13D (1.1%), CAPN3 (1.1%), and PKHD1 (1.1%). There were 488 women with fragile X syndrome-FMR1 gene detection, and 6 patients (1.2%) had FMR1 gene mutation. A total of 426 patients were screened for spinal muscular atrophy-SMN1, and the carrier rate was 3.5%, and the detection rate of parents' co-carrier was 0.5%. CONCLUSION: Monogenic recessive hereditary diseases had a high carrier rate in the population. Pre-pregnancy screening could provide good prenatal and postnatal care guidance for patients and preimplantation genetic testing for monogenic/single gene disorders (PGT-M) and prenatal diagnosis could provide more precise reproductive choices for high-risk parents.


Subject(s)
Genetic Testing , Muscular Atrophy, Spinal , Pregnancy , Humans , Female , Retrospective Studies , Genetic Testing/methods , Prenatal Diagnosis/methods , Mutation , Muscular Atrophy, Spinal/genetics , Fragile X Mental Retardation Protein/genetics , Oxidoreductases/genetics , Membrane Proteins/genetics
13.
Mol Biol Rep ; 51(1): 480, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38578387

ABSTRACT

Fragile X syndrome (FXS) is a genetic disorder characterized by mutation in the FMR1 gene, leading to the absence or reduced levels of fragile X Messenger Ribonucleoprotein 1 (FMRP). This results in neurodevelopmental deficits, including autistic spectrum conditions. On the other hand, Fragile X-associated tremor/ataxia syndrome (FXTAS) is a distinct disorder caused by the premutation in the FMR1 gene. FXTAS is associated with elevated levels of FMR1 mRNA, leading to neurodegenerative manifestations such as tremors and ataxia.Mounting evidence suggests a link between both syndromes and mitochondrial dysfunction (MDF). In this minireview, we critically examine the intricate relationship between FXS, FXTAS, and MDF, focusing on potential therapeutic avenues to counteract or mitigate their adverse effects. Specifically, we explore the role of mitochondrial cofactors and antioxidants, with a particular emphasis on alpha-lipoic acid (ALA), carnitine (CARN) and Coenzyme Q10 (CoQ10). Findings from this review will contribute to a deeper understanding of these disorders and foster novel therapeutic strategies to enhance patient outcomes.


Subject(s)
Fragile X Syndrome , Mitochondrial Diseases , Humans , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Tremor/drug therapy , Tremor/genetics , Antioxidants/therapeutic use , Ataxia/drug therapy , Ataxia/genetics , Fragile X Mental Retardation Protein/genetics
14.
J Cell Biol ; 223(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38587486

ABSTRACT

ß-Coronaviruses remodel host endomembranes to form double-membrane vesicles (DMVs) as replication organelles (ROs) that provide a shielded microenvironment for viral RNA synthesis in infected cells. DMVs are clustered, but the molecular underpinnings and pathophysiological functions remain unknown. Here, we reveal that host fragile X-related (FXR) family proteins (FXR1/FXR2/FMR1) are required for DMV clustering induced by expression of viral non-structural proteins (Nsps) Nsp3 and Nsp4. Depleting FXRs results in DMV dispersion in the cytoplasm. FXR1/2 and FMR1 are recruited to DMV sites via specific interaction with Nsp3. FXRs form condensates driven by liquid-liquid phase separation, which is required for DMV clustering. FXR1 liquid droplets concentrate Nsp3 and Nsp3-decorated liposomes in vitro. FXR droplets facilitate recruitment of translation machinery for efficient translation surrounding DMVs. In cells depleted of FXRs, SARS-CoV-2 replication is significantly attenuated. Thus, SARS-CoV-2 exploits host FXR proteins to cluster viral DMVs via phase separation for efficient viral replication.


Subject(s)
COVID-19 , Fragile X Mental Retardation Protein , Liposomes , RNA-Binding Proteins , SARS-CoV-2 , Humans , Cell Proliferation , Cluster Analysis , COVID-19/metabolism , COVID-19/virology , Cytoplasm , Fragile X Mental Retardation Protein/metabolism , HeLa Cells , Liposomes/metabolism , Organelles , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/metabolism
15.
Neurobiol Dis ; 195: 106496, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38582333

ABSTRACT

Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent electroencephalographic (EEG) studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced phase locking of sound-evoked gamma oscillations. Similar EEG phenotypes are present in mouse models of FXS, but very little is known about the development of such abnormal responses. In the current study, we employed a 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in male P21 and P91 WT and Fmr1 KO mice. This led to several novel findings. First, P91, but not P21, Fmr1 KO mice have significantly increased resting EEG power in the low- and high-gamma frequency bands. Second, both P21 and P91 Fmr1 KO mice have markedly attenuated inter-trial phase coherence (ITPC) to spectrotemporally dynamic auditory stimuli as well as to 40 Hz and 80 Hz auditory steady-state response (ASSR) stimuli. This suggests abnormal temporal processing from early development that may lead to abnormal speech and language function in FXS. Third, we found hemispheric asymmetry of fast temporal processing in the mouse auditory cortex in WT but not Fmr1 KO mice. Together, these findings define a set of EEG phenotypes in young and adult mice that can serve as translational targets for genetic and pharmacological manipulation in phenotypic rescue studies.


Subject(s)
Electroencephalography , Evoked Potentials, Auditory , Fragile X Mental Retardation Protein , Fragile X Syndrome , Animals , Male , Mice , Acoustic Stimulation , Biomarkers , Disease Models, Animal , Electroencephalography/methods , Evoked Potentials, Auditory/physiology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype
16.
Cell Rep ; 43(4): 114068, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38614085

ABSTRACT

The precise anatomical degree of brain X chromosome inactivation (XCI) that is sufficient to alter X-linked disorders in females is unclear. Here, we quantify whole-brain XCI at single-cell resolution to discover a prevalent activation ratio of maternal to paternal X at 60:40 across all divisions of the adult brain. This modest, non-random XCI influences X-linked disease penetrance: maternal transmission of the fragile X mental retardation 1 (Fmr1)-knockout (KO) allele confers 55% of total brain cells with mutant X-active, which is sufficient for behavioral penetrance, while 40% produced from paternal transmission is tolerated. Local XCI mosaicism within affected maternal Fmr1-KO mice further specifies sensorimotor versus social anxiety phenotypes depending on which distinct brain circuitry is most affected, with only a 50%-55% mutant X-active threshold determining penetrance. Thus, our results define a model of X-linked disease penetrance in females whereby distributed XCI among single cells populating brain circuitries can regulate the behavioral penetrance of an X-linked mutation.


Subject(s)
Brain , Mice, Knockout , Penetrance , X Chromosome Inactivation , X Chromosome Inactivation/genetics , Animals , Female , Mice , Brain/metabolism , Male , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Behavior, Animal , Mice, Inbred C57BL , Mosaicism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology
17.
Sensors (Basel) ; 24(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38676203

ABSTRACT

FXTAS is a neurodegenerative disorder occurring in some Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene premutation carriers (PMCs) and is characterized by cerebellar ataxia, tremor, and cognitive deficits that negatively impact balance and gait and increase fall risk. Dual-tasking (DT) cognitive-motor paradigms and challenging balance conditions may have the capacity to reveal markers of FXTAS onset. Our objectives were to determine the impact of dual-tasking and sensory and stance manipulation on balance in FXTAS and potentially detect subtle postural sway deficits in FMR1 PMCs who are asymptomatic for signs of FXTAS on clinical exam. Participants with FXTAS, PMCs without FXTAS, and controls underwent balance testing using an inertial sensor system. Stance, vision, surface stability, and cognitive demand were manipulated in 30 s trials. FXTAS participants had significantly greater total sway area, jerk, and RMS sway than controls under almost all balance conditions but were most impaired in those requiring vestibular control. PMCs without FXTAS had significantly greater RMS sway compared with controls in the feet apart, firm, single task conditions both with eyes open and closed (EC) and the feet together, firm, EC, DT condition. Postural sway deficits in the RMS postural sway variability domain in asymptomatic PMCs might represent prodromal signs of FXTAS. This information may be useful in providing sensitive biomarkers of FXTAS onset and as quantitative balance measures in future interventional trials and longitudinal natural history studies.


Subject(s)
Ataxia , Fragile X Syndrome , Postural Balance , Tremor , Humans , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Tremor/genetics , Tremor/physiopathology , Postural Balance/physiology , Male , Middle Aged , Female , Ataxia/genetics , Ataxia/physiopathology , Aged , Biomarkers , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Adult , Prodromal Symptoms
18.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38627066

ABSTRACT

Autism spectrum disorder (ASD) is often associated with social communication impairments and specific sound processing deficits, for example, problems in following speech in noisy environments. To investigate underlying neuronal processing defects located in the auditory cortex (AC), we performed two-photon Ca2+ imaging in FMR1 (fragile X messenger ribonucleoprotein 1) knock-out (KO) mice, a model for fragile X syndrome (FXS), the most common cause of hereditary ASD in humans. For primary AC (A1) and the anterior auditory field (AAF), topographic frequency representation was less ordered compared with control animals. We additionally analyzed ensemble AC activity in response to various sounds and found subfield-specific differences. In A1, ensemble correlations were lower in general, while in secondary AC (A2), correlations were higher in response to complex sounds, but not to pure tones. Furthermore, sound specificity of ensemble activity was decreased in AAF. Repeating these experiments 1 week later revealed no major differences regarding representational drift. Nevertheless, we found subfield- and genotype-specific changes in ensemble correlation values between the two times points, hinting at alterations in network stability in FMR1 KO mice. These detailed insights into AC network activity and topography in FMR1 KO mice add to the understanding of auditory processing defects in FXS.


Subject(s)
Auditory Cortex , Disease Models, Animal , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Animals , Auditory Cortex/physiopathology , Fragile X Syndrome/physiopathology , Fragile X Syndrome/genetics , Fragile X Mental Retardation Protein/genetics , Male , Mice, Inbred C57BL , Acoustic Stimulation , Auditory Perception/physiology , Mice , Calcium/metabolism
19.
Biomolecules ; 14(4)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38672417

ABSTRACT

Silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene and a consequent lack of FMR protein (FMRP) synthesis are associated with fragile X syndrome, one of the most common inherited intellectual disabilities. FMRP is a multifunctional protein that is involved in many cellular functions in almost all subcellular compartments under both normal and cellular stress conditions in neuronal and non-neuronal cell types. This is achieved through its trafficking signals, nuclear localization signal (NLS), nuclear export signal (NES), and nucleolar localization signal (NoLS), as well as its RNA and protein binding domains, and it is modulated by various post-translational modifications such as phosphorylation, ubiquitination, sumoylation, and methylation. This review summarizes the recent advances in understanding the interaction networks of FMRP with a special focus on FMRP stress-related functions, including stress granule formation, mitochondrion and endoplasmic reticulum plasticity, ribosome biogenesis, cell cycle control, and DNA damage response.


Subject(s)
Cell Nucleolus , Cytosol , Fragile X Mental Retardation Protein , Fragile X Syndrome , Humans , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Cell Nucleolus/metabolism , Cytosol/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Animals , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Protein Processing, Post-Translational
20.
Nat Commun ; 15(1): 3583, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678030

ABSTRACT

Fragile X syndrome (FXS) is an inherited form of intellectual disability caused by the loss of the mRNA-binding fragile X mental retardation protein (FMRP). FXS is characterized by neuronal hyperexcitability and behavioral defects, however the mechanisms underlying these critical dysfunctions remain unclear. Here, using male Fmr1 knockout mouse model of FXS, we identify abnormal extracellular potassium homeostasis, along with impaired potassium channel Kir4.1 expression and function in astrocytes. Further, we reveal that Kir4.1 mRNA is a binding target of FMRP. Finally, we show that the deficit in astroglial Kir4.1 underlies neuronal hyperexcitability and several behavioral defects in Fmr1 knockout mice. Viral delivery of Kir4.1 channels specifically to hippocampal astrocytes from Fmr1 knockout mice indeed rescues normal astrocyte potassium uptake, neuronal excitability, and cognitive and social performance. Our findings uncover an important role for astrocyte dysfunction in the pathophysiology of FXS, and identify Kir4.1 channel as a potential therapeutic target for FXS.


Subject(s)
Astrocytes , Fragile X Mental Retardation Protein , Fragile X Syndrome , Neurons , Potassium Channels, Inwardly Rectifying , Animals , Male , Mice , Astrocytes/metabolism , Behavior, Animal , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Hippocampus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/physiology , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...