Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.035
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Cell Rep ; 43(3): 113906, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38451812

ABSTRACT

Kinesin 1 (KIF5) is one major type of motor protein in neurons, but its members' function in the intact brain remains less studied. Using in vivo two-photon imaging, we find that conditional knockout of Kif5b (KIF5B cKO) in CaMKIIα-Cre-expressing neurons shows heightened turnover and lower stability of dendritic spines in layer 2/3 pyramidal neurons with reduced spine postsynaptic density protein 95 acquisition in the mouse cortex. Furthermore, the RNA-binding protein fragile X mental retardation protein (FMRP) is translocated to the proximity of newly formed spines several hours before the spine formation events in vivo in control mice, but this preceding transport of FMRP is abolished in KIF5B cKO mice. We further find that FMRP is localized closer to newly formed spines after fear extinction, but this learning-dependent localization is disrupted in KIF5B cKO mice. Our findings provide the crucial in vivo evidence that KIF5B is involved in the dendritic targeting of synaptic proteins that underlies dendritic spine plasticity.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Animals , Mice , Dendritic Spines/metabolism , Extinction, Psychological , Fear , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity
16.
Neurobiol Dis ; 194: 106486, 2024 May.
Article in English | MEDLINE | ID: mdl-38548140

ABSTRACT

Fragile X Syndrome (FXS) is a neurodevelopment disorder characterized by cognitive impairment, behavioral challenges, and synaptic abnormalities, with a genetic basis linked to a mutation in the FMR1 (Fragile X Messenger Ribonucleoprotein 1) gene that results in a deficiency or absence of its protein product, Fragile X Messenger Ribonucleoprotein (FMRP). In recent years, mass spectrometry (MS) - based proteomics has emerged as a powerful tool to uncover the complex molecular landscape underlying FXS. This review provides a comprehensive overview of the proteomics studies focused on FXS, summarizing key findings with an emphasis on dysregulated proteins associated with FXS. These proteins span a wide range of cellular functions including, but not limited to, synaptic plasticity, RNA translation, and mitochondrial function. The work conducted in these proteomic studies provides a more holistic understanding to the molecular pathways involved in FXS and considerably enhances our knowledge into the synaptic dysfunction seen in FXS.


Subject(s)
Fragile X Syndrome , Humans , Fragile X Syndrome/genetics , Fragile X Syndrome/therapy , Fragile X Syndrome/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Proteomics , Gene Expression Regulation
17.
Genes (Basel) ; 15(3)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38540415

ABSTRACT

Fragile X syndrome (FXS) is the most common heritable cause of intellectual disability and autism spectrum disorder. The syndrome is often caused by greatly reduced or absent protein expression from the fragile X messenger ribonucleoprotein 1 (FMR1) gene due to expansion of a 5'-non-coding trinucleotide (CGG) element beyond 200 repeats (full mutation). To better understand the complex relationships among FMR1 allelotype, methylation status, mRNA expression, and FMR1 protein (FMRP) levels, FMRP was quantified in peripheral blood mononuclear cells for a large cohort of FXS (n = 154) and control (n = 139) individuals using time-resolved fluorescence resonance energy transfer. Considerable size and methylation mosaicism were observed among individuals with FXS, with FMRP detected only in the presence of such mosaicism. No sample with a minimum allele size greater than 273 CGG repeats had significant levels of FMRP. Additionally, an association was observed between FMR1 mRNA and FMRP levels in FXS samples, predominantly driven by those with the lowest FMRP values. This study underscores the complexity of FMR1 allelotypes and FMRP expression and prompts a reevaluation of FXS therapies aimed at reactivating large full mutation alleles that are likely not capable of producing sufficient FMRP to improve cognitive function.


Subject(s)
Autism Spectrum Disorder , Fragile X Syndrome , Humans , Fragile X Syndrome/genetics , Trinucleotide Repeat Expansion/genetics , Leukocytes, Mononuclear/metabolism , Autism Spectrum Disorder/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
J Mol Diagn ; 26(6): 498-509, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522837

ABSTRACT

Fragile X syndrome (FXS) is the most common heritable form of intellectual disability and is caused by CGG repeat expansions exceeding 200 (full mutation). Such expansions lead to hypermethylation and transcriptional silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene. As a consequence, little or no FMR1 protein (FMRP) is produced; absence of the protein, which normally is responsible for neuronal development and maintenance, causes the syndrome. Previous studies have demonstrated the causal relationship between FMRP levels and cognitive abilities in peripheral blood mononuclear cells (PBMCs) and dermal fibroblast cell lines of patients with FXS. However, it is arguable whether PBMCs or fibroblasts would be the preferred surrogate for measuring molecular markers, particularly FMRP, to represent the cognitive impairment, a core symptom of FXS. To address this concern, CGG repeats, methylation status, FMR1 mRNA, and FMRP levels were measured in both PBMCs and fibroblasts derived from 66 individuals. The findings indicated a strong association between FMR1 mRNA expression levels and CGG repeat numbers in PBMCs of premutation males after correcting for methylation status. Moreover, FMRP expression levels from both PBMCs and fibroblasts of male participants with a hypermethylated full mutation and with mosaicism demonstrated significant association between the intelligence quotient levels and FMRP levels, suggesting that PBMCs may be preferable for FXS clinical studies, because of their greater accessibility.


Subject(s)
DNA Methylation , Fibroblasts , Fragile X Mental Retardation Protein , Fragile X Syndrome , Leukocytes, Mononuclear , Mutation , Humans , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fibroblasts/metabolism , Leukocytes, Mononuclear/metabolism , Male , Fragile X Syndrome/genetics , Fragile X Syndrome/blood , Fragile X Syndrome/diagnosis , Female , Adult , RNA, Messenger/genetics , Adolescent , Trinucleotide Repeat Expansion/genetics , Young Adult , Intelligence/genetics , Middle Aged , Child
19.
J Clin Invest ; 134(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38426491

ABSTRACT

Fragile X syndrome (FXS), the most common inherited cause of intellectual disability and the single-gene cause of autism, is caused by decreased expression of the fragile X messenger ribonucleoprotein protein (FMRP), a ribosomal-associated RNA-binding protein involved in translational repression. Extensive preclinical work in several FXS animal models supported the therapeutic potential of decreasing metabotropic glutamate receptor (mGluR) signaling to correct translation of proteins related to synaptic plasticity; however, multiple clinical trials failed to show conclusive evidence of efficacy. In this issue of the JCI, Berry-Kravis and colleagues conducted the FXLEARN clinical trial to address experimental design concerns from previous trials. Unfortunately, despite treatment of young children with combined pharmacological and learning interventions for a prolonged period, no efficacy of blocking mGluR activity was observed. Future systematic evaluation of potential therapeutic approaches should evaluate consistency between human and animal pathophysiological mechanisms, utilize innovative clinical trial design from FXLEARN, and incorporate translatable biomarkers.


Subject(s)
Fragile X Syndrome , Intellectual Disability , Receptors, Metabotropic Glutamate , Animals , Child , Humans , Child, Preschool , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/therapeutic use , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Neuronal Plasticity
20.
Neurotoxicol Teratol ; 102: 107331, 2024.
Article in English | MEDLINE | ID: mdl-38301979

ABSTRACT

Bisphenol F (BPF) is a potential neurotoxicant used as a replacement for bisphenol A (BPA) in polycarbonate plastics and epoxy resins. We investigated the neurodevelopmental impacts of BPF exposure using Drosophila melanogaster as a model. Our transcriptomic analysis indicated that developmental exposure to BPF caused the downregulation of neurodevelopmentally relevant genes, including those associated with synapse formation and neuronal projection. To investigate the functional outcome of BPF exposure, we evaluated neurodevelopmental impacts across two genetic strains of Drosophila- w1118 (control) and the Fragile X Syndrome (FXS) model-by examining both behavioral and neuronal phenotypes. We found that BPF exposure in w1118 Drosophila caused hypoactive larval locomotor activity, decreased time spent grooming by adults, reduced courtship activity, and increased the severity but not frequency of ß-lobe midline crossing defects by axons in the mushroom body. In contrast, although BPF reduced peristaltic contractions in FXS larvae, it had no impact on other larval locomotor phenotypes, grooming activity, or courtship activity. Strikingly, BPF exposure reduced both the severity and frequency of ß-lobe midline crossing defects in the mushroom body of FXS flies, a phenotype previously observed in FXS flies exposed to BPA. This data indicates that BPF can affect neurodevelopment and its impacts vary depending on genetic background. Further, BPF may elicit a gene-environment interaction with Drosophila fragile X messenger ribonucleoprotein 1 (dFmr1)-the ortholog of human FMR1, which causes fragile X syndrome and is the most common monogenetic cause of intellectual disability and autism spectrum disorder.


Subject(s)
Autism Spectrum Disorder , Drosophila Proteins , Fragile X Syndrome , Phenols , Animals , Humans , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Fragile X Syndrome/chemically induced , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Autism Spectrum Disorder/metabolism , Mushroom Bodies/metabolism , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Drosophila , Benzhydryl Compounds/toxicity , Gene Expression
SELECTION OF CITATIONS
SEARCH DETAIL
...