Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.037
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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 , Deep Brain Stimulation/methods , Male , Fragile X Syndrome/therapy , Tremor/therapy , Ataxia/therapy , Magnetic Resonance Imaging , Fragile X Mental Retardation Protein/genetics , Adult , Middle Aged
7.
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
8.
Neurosci Biobehav Rev ; 161: 105688, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670298

ABSTRACT

Pyramidal neurons have a pivotal role in the cognitive capabilities of neocortex. Though they have been predominantly modeled as integrate-and-fire point processors, many of them have another point of input integration in their apical dendrites that is central to mechanisms endowing them with the sensitivity to context that underlies basic cognitive capabilities. Here we review evidence implicating impairments of those mechanisms in three major neurodevelopmental disabilities, fragile X, Down syndrome, and fetal alcohol spectrum disorders. Multiple dysfunctions of the mechanisms by which pyramidal cells are sensitive to context are found to be implicated in all three syndromes. Further deciphering of these cellular mechanisms would lead to the understanding of and therapies for learning disabilities beyond any that are currently available.


Subject(s)
Learning Disabilities , Humans , Animals , Learning Disabilities/physiopathology , Learning Disabilities/etiology , Pyramidal Cells/physiology , Fetal Alcohol Spectrum Disorders/physiopathology , Neurodevelopmental Disorders/physiopathology , Down Syndrome/physiopathology , Fragile X Syndrome/physiopathology
9.
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
10.
Biochim Biophys Acta Proteins Proteom ; 1872(4): 141019, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38641086

ABSTRACT

The Fragile X messenger ribonucleoprotein (FMRP) is a multi-domain protein involved in interactions with various macromolecules, including proteins and coding/non-coding RNAs. The three KH domains (KH0, KH1 and KH2) within FMRP are recognized for their roles in mRNA binding. In the context of Fragile X syndrome (FXS), over-and-above CGG triplet repeats expansion, three specific point mutations have been identified, each affecting one of the three KH domains (R138QKH0, G266EKH1, and I304NKH2) resulting in the expression of non-functional FMRP. This study aims to elucidate the molecular mechanism underlying the loss of function associated with the G266EKH1 pathological variant. We investigate the conformational and dynamic properties of the isolated KH1 domain and the two KH1 site-directed mutants G266EKH1 and G266AKH1. Employing a combined in vitro and in silico approach, we reveal that the G266EKH1 variant lacks the characteristic features of a folded domain. This observation provides an explanation for functional impairment observed in FMRP carrying the G266E mutation within the KH1 domain, as it renders the domain unable to fold properly. Molecular Dynamics simulations suggest a pivotal role for residue 266 in regulating the structural stability of the KH domains, primarily through stabilizing the α-helices of the domain. Overall, these findings enhance our comprehension of the molecular basis for the dysfunction associated with the G266EKH1 variant in FMRP.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/chemistry , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Humans , Protein Domains , Molecular Dynamics Simulation , Protein Conformation , Mutagenesis, Site-Directed
11.
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
12.
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
13.
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
14.
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
15.
Cell Rep ; 43(5): 114124, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38630591

ABSTRACT

High-penetrance mutations affecting mental health can involve genes ubiquitously expressed in the brain. Whether the specific patterns of dysfunctions result from ubiquitous circuit deficits or might reflect selective vulnerabilities of targetable subnetworks has remained unclear. Here, we determine how loss of ubiquitously expressed fragile X mental retardation protein (FMRP), the cause of fragile X syndrome, affects brain networks in Fmr1y/- mice. We find that in wild-type mice, area-specific knockout of FMRP in the adult mimics behavioral consequences of area-specific silencing. By contrast, the functional axis linking the ventral hippocampus (vH) to the prelimbic cortex (PreL) is selectively affected in constitutive Fmr1y/- mice. A chronic alteration in late-born parvalbumin interneuron networks across the vH-PreL axis rescued by VIP signaling specifically accounts for deficits in vH-PreL theta-band network coherence, ensemble assembly, and learning functions of Fmr1y/- mice. Therefore, vH-PreL axis function exhibits a selective vulnerability to loss of FMRP in the vH or PreL, leading to learning and memory dysfunctions in fragile X mice.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Hippocampus , Interneurons , Parvalbumins , Animals , Parvalbumins/metabolism , Interneurons/metabolism , Hippocampus/metabolism , Mice , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Fragile X Syndrome/pathology , Mice, Knockout , Male , Mice, Inbred C57BL , Learning/physiology , Nerve Net/metabolism , Nerve Net/physiopathology , Nerve Net/pathology
16.
J Neurosci ; 44(21)2024 May 22.
Article in English | MEDLINE | ID: mdl-38664011

ABSTRACT

Fragile X syndrome (FXS) arises from the loss of fragile X messenger ribonucleoprotein (FMRP) needed for normal neuronal excitability and circuit functions. Recent work revealed that FMRP contributes to mossy fiber long-term potentiation by adjusting the Kv4 A-type current availability through interactions with a Cav3-Kv4 ion channel complex, yet the mechanism has not yet been defined. In this study using wild-type and Fmr1 knock-out (KO) tsA-201 cells and cerebellar sections from male Fmr1 KO mice, we show that FMRP associates with all subunits of the Cav3.1-Kv4.3-KChIP3 complex and is critical to enabling calcium-dependent shifts in Kv4.3 inactivation to modulate the A-type current. Specifically, upon depolarization Cav3 calcium influx activates dual-specific phosphatase 1/6 (DUSP1/6) to deactivate ERK1/2 (ERK) and lower phosphorylation of Kv4.3, a signaling pathway that does not function in Fmr1 KO cells. In Fmr1 KO mouse tissue slices, cerebellar granule cells exhibit a hyperexcitable response to membrane depolarizations. Either incubating Fmr1 KO cells or in vivo administration of a tat-conjugated FMRP N-terminus fragment (FMRP-N-tat) rescued Cav3-Kv4 function and granule cell excitability, with a decrease in the level of DUSP6. Together these data reveal a Cav3-activated DUSP signaling pathway critical to the function of a FMRP-Cav3-Kv4 complex that is misregulated in Fmr1 KO conditions. Moreover, FMRP-N-tat restores function of this complex to rescue calcium-dependent control of neuronal excitability as a potential therapeutic approach to alleviating the symptoms of FXS.


Subject(s)
Calcium , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neurons , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Mice , Male , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Neurons/metabolism , Calcium/metabolism , Mice, Inbred C57BL , Shal Potassium Channels/metabolism , Shal Potassium Channels/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
17.
Pharmacol Res ; 203: 107176, 2024 May.
Article in English | MEDLINE | ID: mdl-38583687

ABSTRACT

Cannabidiol (CBD), a non-psychotomimetic constituent of Cannabis sativa, has been recently approved for epileptic syndromes often associated with Autism spectrum disorder (ASD). However, the putative efficacy and mechanism of action of CBD in patients suffering from ASD and related comorbidities remain debated, especially because of the complex pharmacology of CBD. We used pharmacological, immunohistochemical and biochemical approaches to investigate the effects and mechanisms of action of CBD in the recently validated Fmr1-Δexon 8 rat model of ASD, that is also a model of Fragile X Syndrome (FXS), the leading monogenic cause of autism. CBD rescued the cognitive deficits displayed by juvenile Fmr1-Δexon 8 animals, without inducing tolerance after repeated administration. Blockade of CA1 hippocampal GPR55 receptors prevented the beneficial effect of both CBD and the fatty acid amide hydrolase (FAAH) inhibitor URB597 in the short-term recognition memory deficits displayed by Fmr1-Δexon 8 rats. Thus, CBD may exert its beneficial effects through CA1 hippocampal GPR55 receptors. Docking analysis further confirmed that the mechanism of action of CBD might involve competition for brain fatty acid binding proteins (FABPs) that deliver anandamide and related bioactive lipids to their catabolic enzyme FAAH. These findings demonstrate that CBD reduced cognitive deficits in a rat model of FXS and provide initial mechanistic insights into its therapeutic potential in neurodevelopmental disorders.


Subject(s)
Cannabidiol , Disease Models, Animal , Fragile X Syndrome , Hippocampus , Receptors, Cannabinoid , Recognition, Psychology , Animals , Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Receptors, Cannabinoid/metabolism , Male , Recognition, Psychology/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Memory/drug effects , Receptors, G-Protein-Coupled/metabolism , Molecular Docking Simulation
18.
Res Dev Disabil ; 148: 104719, 2024 May.
Article in English | MEDLINE | ID: mdl-38507982

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability. Early Check, a voluntary newborn screening study, screened 18,833 newborns for FXS over ∼3 years. Exploring parental attitudes and perspectives can provide insight to the potential future acceptability of public health screening. METHODS AND PROCEDURES: Mothers of infants who received a screen positive result for FXS (n = 6) or fragile X premutation (FXPM; n = 18) were interviewed about their perceptions and experiences. OUTCOMES AND RESULTS: Mothers of children with FXS described utility in receiving information about their child, particularly to monitor for potential developmental issues and intervene early; overall mothers did not regret participating. Mothers reported various reactions to receiving the FXS or FXPM results including (1) stress and worry; (2) guilt; (3) sadness and disappointment; (4) neutrality, relief, and acceptance; and (5) confusion and uncertainty. CONCLUSIONS AND IMPLICATIONS: Despite initial reactions such as sadness, stress, and worry, mothers found value in learning of their child's presymptomatic diagnosis of FXS, particularly the anticipated long-term benefits of early diagnosis to their child's health and wellbeing. Our results indicate that professionals returning positive newborn screening results should anticipate and prepare for reactions such as parental shock, guilt, sadness, and uncertainty. Genetic counseling and psychosocial support are critical to supporting families.


Subject(s)
Fragile X Syndrome , Intellectual Disability , Female , Infant , Child , Humans , Infant, Newborn , Fragile X Syndrome/psychology , Neonatal Screening , Genetic Testing , Intellectual Disability/genetics , Parents
19.
Genes (Basel) ; 15(3)2024 Mar 03.
Article in English | MEDLINE | ID: mdl-38540390

ABSTRACT

Fragile X syndrome (FXS) is the leading inherited cause of intellectual disability (ID) and single gene cause of autism. Although most patients with FXS and the full mutation (FM) have complete methylation of the fragile X messenger ribonucleoprotein 1 (FMR1) gene, some have mosaicism in methylation and/or CGG repeat size, and few have completely unmethylated FM alleles. Those with a complete lack of methylation are rare, with little literature about the cognitive and behavioral phenotypes of these individuals. A review of past literature was conducted regarding individuals with unmethylated and mosaic FMR1 FM. We report three patients with an unmethylated FM FMR1 alleles without any behavioral or cognitive deficits. This is an unusual presentation for men with FM as most patients with an unmethylated FM and no behavioral phenotypes do not receive fragile X DNA testing or a diagnosis of FXS. Our cases showed that mosaic males with unmethylated FMR1 FM alleles may lack behavioral phenotypes due to the presence of smaller alleles producing the FMR1 protein (FMRP). However, these individuals could be at a higher risk of developing fragile X-associated tremor/ataxia syndrome (FXTAS) due to the increased expression of mRNA, similar to those who only have a premutation.


Subject(s)
Ataxia , Fragile X Syndrome , Tremor , Male , Humans , Fragile X Syndrome/genetics , Fragile X Syndrome/complications , DNA Methylation/genetics , Fragile X Mental Retardation Protein/genetics , Mutation
20.
Genesis ; 62(2): e23590, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490949

ABSTRACT

The role of neurogenesis in neurodevelopmental disorders (NDDs) merits much attention. The complex process by which stem cells produce daughter cells that in turn differentiate into neurons, migrate various distances, and form synaptic connections that are then refined by neuronal activity or experience is integral to the development of the nervous system. Given the continued postnatal neurogenesis that occurs in the mammalian olfactory system, it provides an ideal model for understanding how disruptions in distinct stages of neurogenesis contribute to the pathophysiology of various NDDs. This review summarizes and discusses what is currently known about the disruption of neurogenesis within the olfactory system as it pertains to attention-deficit/hyperactivity disorder, autism spectrum disorder, Down syndrome, Fragile X syndrome, and Rett syndrome. Studies included in this review used either human subjects, mouse models, or Drosophila models, and lay a compelling foundation for continued investigation of NDDs by utilizing the olfactory system.


Subject(s)
Autism Spectrum Disorder , Fragile X Syndrome , Neurodevelopmental Disorders , Mice , Animals , Humans , Neurogenesis/physiology , Fragile X Syndrome/genetics , Neurons , Neurodevelopmental Disorders/genetics , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL
...