4,006 results on '"Fragile X Syndrome genetics"'
Search Results
252. Reversal of behavioural phenotype by the cannabinoid-like compound VSN16R in fragile X syndrome mice.
- Author
-
Hurley MJ, Deacon RMJ, Chan AWE, Baker D, Selwood DL, and Cogram P
- Subjects
- Animals, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Humans, Mice, Phenotype, Autism Spectrum Disorder, Cannabinoids pharmacology, Cannabinoids therapeutic use, Fragile X Syndrome drug therapy, Fragile X Syndrome genetics
- Abstract
Fragile X syndrome is the most common inherited intellectual disability and mono-genetic cause of autism spectrum disorder. It is a neurodevelopmental condition occurring due to a CGG trinucleotide expansion in the FMR1 gene. Polymorphisms and variants in large-conductance calcium-activated potassium channels are increasingly linked to intellectual disability and loss of FMR protein causes reduced large-conductance calcium-activated potassium channel activity leading to abnormalities in synapse function. Using the cannabinoid-like large-conductance calcium-activated potassium channel activator VSN16R we rescued behavioural deficits such as repetitive behaviour, hippocampal dependent tests of daily living, hyperactivity and memory in a mouse model of fragile X syndrome. VSN16R has been shown to be safe in a phase 1 study in healthy volunteers and in a phase 2 study in patients with multiple sclerosis with high oral bioavailability and no serious adverse effects reported. VSN16R could therefore be directly utilized in a fragile X syndrome clinical study. Moreover, VSN16R showed no evidence of tolerance, which strongly suggests that chronic VSN16R may have great therapeutic value for fragile X syndrome and autism spectrum disorder. This study provides new insight into the pathophysiology of fragile X syndrome and identifies a new pathway for drug intervention for this debilitating disorder., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
253. Phosphodiesterase 2A inhibition corrects the aberrant behavioral traits observed in genetic and environmental preclinical models of Autism Spectrum Disorder.
- Author
-
Schiavi S, Carbone E, Melancia F, di Masi A, Jarjat M, Brau F, Cardarelli S, Giorgi M, Bardoni B, and Trezza V
- Subjects
- Animals, Cyclic Nucleotide Phosphodiesterases, Type 2 metabolism, Female, Fragile X Mental Retardation Protein, Pregnancy, Rats, Valproic Acid pharmacology, Autism Spectrum Disorder chemically induced, Autism Spectrum Disorder drug therapy, Autism Spectrum Disorder genetics, Fragile X Syndrome genetics
- Abstract
Pharmacological inhibition of phosphodiesterase 2A (PDE2A), which catalyzes the hydrolysis of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), has recently been proposed as a novel therapeutic tool for Fragile X Syndrome (FXS), the leading monogenic cause of Autism Spectrum Disorder (ASD). Here, we investigated the role of PDE2A in ASD pathogenesis using two rat models that reflect one of either the genetic or environmental factors involved in the human disease: the genetic Fmr1-
Δ exon 8 rat model and the environmental rat model based on prenatal exposure to valproic acid (VPA, 500 mg/kg). Prior to behavioral testing, the offspring was treated with the PDE2A inhibitor BAY607550 (0.05 mg/kg at infancy, 0.1 mg/kg at adolescence and adulthood). Socio-communicative symptoms were assessed in both models through the ultrasonic vocalization test at infancy and three-chamber test at adolescence and adulthood, while cognitive impairments were assessed by the novel object recognition test in Fmr1-Δ exon 8 rats (adolescence and adulthood) and by the inhibitory avoidance test in VPA-exposed rats (adulthood). PDE2A enzymatic activity in VPA-exposed infant rats was also assessed. In line with the increased PDE2A enzymatic activity previously observed in the brain of Fmr1-KO animals, we found an altered upstream regulation of PDE2A activity in the brain of VPA-exposed rats at an early developmental age (p < 0.05). Pharmacological inhibition of PDE2A normalized the communicative (p < 0.01, p < 0.05), social (p < 0.001, p < 0.05), and cognitive impairment (p < 0.001) displayed by both Fmr1-Δ exon 8 and VPA-exposed rats. Altogether, these data highlight a key role of PDE2A in brain development and point to PDE2A inhibition as a promising pharmacological approach for the deficits common to both FXS and ASD., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
254. Identification of microRNAs associated with human fragile X syndrome using next-generation sequencing.
- Author
-
Sotoudeh Anvari M, Vasei H, Najmabadi H, Badv RS, Golipour A, Mohammadi-Yeganeh S, Salehi S, Mohamadi M, Goodarzynejad H, and Mowla SJ
- Subjects
- Biomarkers, Female, Fragile X Mental Retardation Protein genetics, High-Throughput Nucleotide Sequencing, Humans, Iran, Male, Fragile X Syndrome genetics, MicroRNAs metabolism
- Abstract
Fragile X syndrome (FXS) is caused by a mutation in the FMR1 gene which can lead to a loss or shortage of the FMR1 protein. This protein interacts with specific miRNAs and can cause a range of neurological disorders. Therefore, miRNAs could act as a novel class of biomarkers for common CNS diseases. This study aimed to test this theory by exploring the expression profiles of various miRNAs in Iranian using deep sequencing-based technologies and validating the miRNAs affecting the expression of the FMR1 gene. Blood samples were taken from 15 patients with FXS (9 males, 6 females) and 12 controls. 25 miRNAs were differentially expressed in individuals with FXS compared to controls. Levels of 9 miRNAs were found to be significantly changed (3 upregulated and 6 downregulated). In Patients, the levels of hsa-miR-532-5p, hsa-miR-652-3p and hsa-miR-4797-3p were significantly upregulated while levels of hsa-miR-191-5p, hsa-miR-181-5p, hsa-miR-26a-5p, hsa-miR-30e-5p, hsa-miR-186-5p, and hsa-miR-4797-5p exhibited significant downregulation; and these dysregulations were confirmed by RT-qPCR. This study presents among the first evidence of altered miRNA expression in blood samples from patients with FXS, which could be used for diagnostic, prognostic, and treatment purposes. Larger studies are required to confirm these preliminary results., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
255. FMRP promotes transcription-coupled homologous recombination via facilitating TET1-mediated m5C RNA modification demethylation.
- Author
-
Yang H, Wang Y, Xiang Y, Yadav T, Ouyang J, Phoon L, Zhu X, Shi Y, Zou L, and Lan L
- Subjects
- Cytosine, Demethylation, Homologous Recombination, Humans, Mixed Function Oxygenases metabolism, Proto-Oncogene Proteins metabolism, RNA genetics, RNA metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics
- Abstract
RNA modifications regulate a variety of cellular processes including DNA repair.The RNA methyltransferase TRDMT1 generates methyl-5-cytosine (m5C) on messen-ger RNA (mRNA) at DNA double-strand breaks (DSBs) in transcribed regions, pro-moting transcription-coupled homologous recombination (HR). Here, we identifiedthat Fragile X mental retardation protein (FMRP) promotes transcription-coupled HRvia its interaction with both the m5C writer TRDMT1 and the m5C eraser ten-eleventranslocation protein 1 (TET1). TRDMT1, FMRP, and TET1 function in a temporalorder at the transcriptionally active sites of DSBs. FMRP displays a higher affinity forDNA:RNA hybrids containing m5C-modified RNA than for hybrids without modifica-tion and facilitates demethylation of m5C by TET1 in vitro. Loss of either the chroma-tin- or RNA-binding domain of FMRP compromises demethylation of damage-inducedm5C in cells. Importantly, FMRP is required for R-loop resolving in cells. Due to unre-solved R-loop and m5C preventing completion of DSB repair, FMRP depletion or lowexpression leads to delayed repair of DSBs at transcriptionally active sites and sensitizescancer cells to radiation in a BRCA-independent manner. Together, ourfindings presentan m5C reader, FMRP, which acts as a coordinator between the m5C writer and eraserto promote mRNA-dependent repair and cell survival in cancer.
- Published
- 2022
- Full Text
- View/download PDF
256. The novel potent GSK3 inhibitor AF3581 reverts fragile X syndrome phenotype.
- Author
-
Porceddu PF, Ciampoli M, Romeo E, Garrone B, Durando L, Milanese C, Di Giorgio FP, and Reggiani A
- Subjects
- Animals, Disease Models, Animal, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Glycogen Synthase Kinase 3 genetics, Glycogen Synthase Kinase 3 metabolism, Mice, Mice, Knockout, Phenotype, Fragile X Syndrome drug therapy, Fragile X Syndrome genetics
- Abstract
Glycogen-synthase kinase 3 (GSK3) is a kinase mediating phosphorylation on serine and threonine amino acid residues of several target molecules. The enzyme is involved in the regulation of many cellular processes and aberrant activity of GSK3 has been linked to several disease conditions such as fragile X syndrome (FXS). Recent evidences demonstrating an increased activity of GSK3 in murine models of FXS, suggest that dysregulation/hyperactivation of the GSK3 path should contribute to FXS development. A likely possibility could be that in FXS there is a functional impairment of the upstream inhibitory input over GSK3 thus making overactive the kinase. Since GSK3 signaling is a central regulatory node for critical neurodevelopmental pathways, understanding the contribution of GSK3 dysregulation to FXS, may provide novel targets for therapeutic interventions for this disease. In this study we used AF3581, a potent GSK3 inhibitor that we recently discovered, in an in vivo FXS mouse model to elucidate the crucial role of GSK3 in specific behavioral patterns (locomotor activity, sensorimotor gating and social behavior) associated with this disease. All the behavioral alterations manifested by Fmr1 knockout mice were reverted after a chronic treatment with our GSK3 inhibitor, confirming the importance of this pathway as a therapeutic target., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
257. The Use of "Retardation" in FRAXA, FMRP, FMR1 and Other Designations.
- Author
-
Herring J, Johnson K, and Richstein J
- Subjects
- Humans, Nuclear Proteins metabolism, Poland, RNA-Binding Proteins, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics
- Abstract
The European Fragile X Network met in Wroclaw, Poland, November 2021, and agreed to work towards the eradication of the word "retardation" in regard to the naming of the fragile X gene (FRAXA) and protein (FMRP). There are further genes which have "retardation" or abbreviations for "retardation" in their names or full designations, including FMR1, FMR2, FXR1, FXR2, NUFIP1, AFF1, CYFIP1, etc. "Retardation" was commonly used as a term in years past, but now any reference, even in an abbreviation, is offensive. This article discusses the stigmatisation associated with "retardation", which leads to discrimination; the inaccuracy of using "retardation" in these designations; and the breadth of fragile X syndrome being beyond that of neurodiversity. A more inclusive terminology is called for, one which ceases to use any reference to "retardation". Precedents for offensive gene names being altered is set out. The proposal is to approach the HGNC (HUGO [Human Genome Organisation] Gene Nomenclature Committee) for new terminology to be enacted. Ideas from other researchers in the field are welcomed.
- Published
- 2022
- Full Text
- View/download PDF
258. CMTr cap-adjacent 2'-O-ribose mRNA methyltransferases are required for reward learning and mRNA localization to synapses.
- Author
-
Haussmann IU, Wu Y, Nallasivan MP, Archer N, Bodi Z, Hebenstreit D, Waddell S, Fray R, and Soller M
- Subjects
- Animals, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Reward, Ribose metabolism, Synapses metabolism, Fragile X Syndrome genetics, Methyltransferases genetics, Methyltransferases metabolism
- Abstract
Cap-adjacent nucleotides of animal, protist and viral mRNAs can be O-methylated at the 2' position of the ribose (cOMe). The functions of cOMe in animals, however, remain largely unknown. Here we show that the two cap methyltransferases (CMTr1 and CMTr2) of Drosophila can methylate the ribose of the first nucleotide in mRNA. Double-mutant flies lack cOMe but are viable. Consistent with prominent neuronal expression, they have a reward learning defect that can be rescued by conditional expression in mushroom body neurons before training. Among CMTr targets are cell adhesion and signaling molecules. Many are relevant for learning, and are also targets of Fragile X Mental Retardation Protein (FMRP). Like FMRP, cOMe is required for localization of untranslated mRNAs to synapses and enhances binding of the cap binding complex in the nucleus. Hence, our study reveals a mechanism to co-transcriptionally prime mRNAs by cOMe for localized protein synthesis at synapses., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
259. Fragile X premutation carrier screening in Pakistani preconception women in primary care consultation.
- Author
-
Meraj N, Yasin M, Rehman ZU, Tahir H, Jadoon H, Khan N, Shahid R, Zubair M, Zulfiqar I, Jabeen M, Neelam S, Hameed A, and Saleha S
- Subjects
- Child, Female, Humans, Mutation, Pakistan, Primary Health Care, Referral and Consultation, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome epidemiology, Fragile X Syndrome genetics
- Abstract
Purpose: Women of reproductive age who carry fragile X premutation (PM) alleles have 56 to 200 CGG repeats in the 5'-untranslated region of FMR1 gene are at increased risk for producing children with intellectual disabilities (ID) or autism spectrum disorders (ASD) due to expansion of PM alleles to full mutation alleles (> 200 repeats) during maternal transmission., Methods: In present study fragile X PM carrier screening was performed in total 808 women who were consulting primary health care centers for preconception care in Khyber Pakhtunkhwa region of Pakistan between April, 2018 and December, 2020. Polymerase chain reaction (PCR) was performed for detection of PM carrier women and the CGG repeats number was confirmed by Southern blotting and capillary electrophoresis., Results: The prevalence rate for PM carriers among preconception women was found to be 0.7% that was contributed by 0.5% women in risk group (RG1) with family history of ID and 0.2% in risk group 2 (RG2) with family history of ASD. PM carrier women had at least one affected child or sibling. In addition, the preconception women with FMR1 PM alleles were found to be at increased risk for primary ovary insufficiency (RG1: P = 0.0265, RG2: P = 0.0389), postpartum depression (RG1: P = 0.0240, RG2: P = 0.0501) and neuropsychiatric disorders (RG1: P = 0.0389, RG2: P = 0.0432)., Conclusions: Current study provides first evidence of fragile X PM carrier screening in Pakistani preconception women in primary care consultation. Findings of current study may help to improve preconception care and to reduce burden of fragile X associated disorders in our population., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
260. Restoration of FMRP expression in adult V1 neurons rescues visual deficits in a mouse model of fragile X syndrome.
- Author
-
Yang C, Tian Y, Su F, Wang Y, Liu M, Wang H, Cui Y, Yuan P, Li X, Li A, Gong H, Luo Q, Zhu D, Cao P, Liu Y, Wang X, Luo MH, Xu F, Xiong W, Wang L, Li XY, and Zhang C
- Subjects
- Animals, Disease Models, Animal, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Humans, Mice, Mice, Knockout, Neurons metabolism, Fragile X Syndrome complications, Fragile X Syndrome genetics, Fragile X Syndrome metabolism
- Abstract
Many people affected by fragile X syndrome (FXS) and autism spectrum disorders have sensory processing deficits, such as hypersensitivity to auditory, tactile, and visual stimuli. Like FXS in humans, loss of Fmr1 in rodents also cause sensory, behavioral, and cognitive deficits. However, the neural mechanisms underlying sensory impairment, especially vision impairment, remain unclear. It remains elusive whether the visual processing deficits originate from corrupted inputs, impaired perception in the primary sensory cortex, or altered integration in the higher cortex, and there is no effective treatment. In this study, we used a genetic knockout mouse model (Fmr1
KO ), in vivo imaging, and behavioral measurements to show that the loss of Fmr1 impaired signal processing in the primary visual cortex (V1). Specifically, Fmr1KO mice showed enhanced responses to low-intensity stimuli but normal responses to high-intensity stimuli. This abnormality was accompanied by enhancements in local network connectivity in V1 microcircuits and increased dendritic complexity of V1 neurons. These effects were ameliorated by the acute application of GABAA receptor activators, which enhanced the activity of inhibitory neurons, or by reintroducing Fmr1 gene expression in knockout V1 neurons in both juvenile and young-adult mice. Overall, V1 plays an important role in the visual abnormalities of Fmr1KO mice and it could be possible to rescue the sensory disturbances in developed FXS and autism patients., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
261. Small Molecule Screening Discovers Compounds that Reduce FMRpolyG Protein Aggregates and Splicing Defect Toxicity in Fragile X-Associated Tremor/Ataxia Syndrome.
- Author
-
Verma AK, Khan E, Mishra SK, and Kumar A
- Subjects
- Ataxia metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Humans, Molecular Docking Simulation, Protein Aggregates, Tremor metabolism, Trinucleotide Repeat Expansion genetics, Fragile X Syndrome drug therapy, Fragile X Syndrome genetics, Fragile X Syndrome metabolism
- Abstract
Expansion of CGG trinucleotide repeats in 5' untranslated region of the FMR1 gene is the causative mutation of neurological diseases such as fragile X syndrome (FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), and ovarian disorder such as fragile X-associated primary ovarian insufficiency (FXPOI). CGG repeats containing FMR1 transcripts form the toxic ribonuclear aggregates, abrupt pre-mRNA splicing, and cause repeat-associated non-AUG translation, leading to the disease symptoms. Here, we utilized a small molecule library of ~ 250,000 members obtained from the National Cancer Institute (NCI) and implemented a shape-based screening approach to identify the candidate small molecules that mitigate toxic CGG RNA-mediated pathogenesis. The compounds obtained from screening were further assessed for their affinity and selectivity towards toxic CGG repeat RNA by employing fluorescence-binding experiment and isothermal calorimetry titration assay. Three candidate molecules B1, B4, and B11 showed high affinity and selectivity for expanded CGG repeats RNA. Further, NMR spectroscopy, gel mobility shift assay, CD spectroscopy, UV-thermal denaturation assay, and molecular docking affirmed their high affinity and selectivity for toxic CGG RNAs. Next, these lead compounds selectively improved the pre-mRNA alternative splicing defects with no perturbation in global splicing efficacy and simultaneously reduced the FMR1polyG protein aggregate formation without affecting the downstream expression of the gene. Taken together these findings, we addressed compound B1, B4, and B11 as potential lead molecules for developing promising therapeutics against FXTAS. Herein, this study, we have utilized shape similarity approach to screen the NCI library and found out the potential candidate which improves the pre-mRNA splicing defects and reduces FMR1polyG aggregations., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
262. Expression of FMRpolyG in Peripheral Blood Mononuclear Cells of Women with Fragile X Mental Retardation 1 Gene Premutation.
- Author
-
Nguyen XP, Vilkaite A, Messmer B, Dietrich JE, Hinderhofer K, Schäkel K, Strowitzki T, and Rehnitz J
- Subjects
- Ataxia genetics, Ataxia metabolism, Case-Control Studies, Female, Fragile X Mental Retardation Protein genetics, Humans, Leukocytes, Mononuclear metabolism, Tremor genetics, Tremor metabolism, Fragile X Syndrome genetics, Intellectual Disability genetics
- Abstract
Fragile X-associated primary ovarian insufficiency (FXPOI) is characterized by oligo/amenorrhea and hypergonadotropic hypogonadism and is caused by the expansion of the CGG repeat in the 5'UTR of Fragile X Mental Retardation 1 ( FMR1) . Approximately 20% of women carrying an FMR1 premutation (PM) allele (55-200 CGG repeat) develop FXPOI. Repeat Associated Non-AUG (RAN)-translation dependent on the variable CGG-repeat length is thought to cause FXPOI, due to the production of a polyglycine-containing FMR1 protein, FMRpolyG. Peripheral blood monocyte cells (PBMCs) and granulosa cells (GCs) were collected to detect FMRpolyG and its cell type-specific expression in FMR1 PM carriers by immunofluorescence staining (IF), Western blotting (WB), and flow cytometric analysis (FACS). For the first time, FMRpolyG aggregates were detected as ubiquitin-positive inclusions in PBMCs from PM carriers, whereas only a weak signal without inclusions was detected in the controls. The expression pattern of FMRpolyG in GCs was comparable to that in the lymphocytes. We detected FMRpolyG as a 15- to 25-kDa protein in the PBMCs from two FMR1 PM carriers, with 124 and 81 CGG repeats. Flow cytometric analysis revealed that FMRpolyG was significantly higher in the T cells from PM carriers than in those from non-PM carriers. The detection of FMRpolyG aggregates in the peripheral blood and granulosa cells of PM carriers suggests that it may have a toxic potential and an immunological role in ovarian damage in the development of FXPOI.
- Published
- 2022
- Full Text
- View/download PDF
263. #ESHREjc report: Catch 22-is PGT a number game? Efficacy of PGT and the importance of counselling.
- Author
-
Fraire-Zamora JJ, Serdarogullari M, Kohlhepp F, Sharma K, Popovic M, Pujol A, and Liperis G
- Subjects
- Counseling, Female, Fragile X Mental Retardation Protein genetics, Genetic Testing, Humans, Pregnancy, Fragile X Syndrome genetics, Preimplantation Diagnosis
- Published
- 2022
- Full Text
- View/download PDF
264. Correspondence on "Artificial intelligence-assisted phenotype discovery of fragile X syndrome in a population-based sample" by Movaghar et al.
- Author
-
Ménard T
- Subjects
- Artificial Intelligence, Fragile X Mental Retardation Protein genetics, Humans, Phenotype, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics
- Abstract
Competing Interests: Conflict of Interest Timothé Ménard was employed by F. Hoffmann-La Roche (Roche) at the time this article was completed. However, this article has been written independently from the author's employment.
- Published
- 2022
- Full Text
- View/download PDF
265. Neurodegenerative diseases associated with non-coding CGG tandem repeat expansions.
- Author
-
Zhou ZD, Jankovic J, Ashizawa T, and Tan EK
- Subjects
- Ataxia genetics, Ataxia pathology, Humans, Tremor genetics, Tremor pathology, Trinucleotide Repeat Expansion genetics, Fragile X Syndrome genetics, Fragile X Syndrome pathology, Neurodegenerative Diseases genetics, Neurodegenerative Diseases pathology
- Abstract
Non-coding CGG repeat expansions cause multiple neurodegenerative disorders, including fragile X-associated tremor/ataxia syndrome, neuronal intranuclear inclusion disease, oculopharyngeal myopathy with leukodystrophy, and oculopharyngodistal myopathy. The underlying genetic causes of several of these diseases have been identified only in the past 2-3 years. These expansion disorders have substantial overlapping clinical, neuroimaging and histopathological features. The shared features suggest common mechanisms that could have implications for the development of therapies for this group of diseases - similar therapeutic strategies or drugs may be effective for various neurodegenerative disorders induced by non-coding CGG expansions. In this Review, we provide an overview of clinical and pathological features of these CGG repeat expansion diseases and consider the likely pathological mechanisms, including RNA toxicity, CGG repeat-associated non-AUG-initiated translation, protein aggregation and mitochondrial impairment. We then discuss future research needed to improve the identification and diagnosis of CGG repeat expansion diseases, to improve modelling of these diseases and to understand their pathogenesis. We also consider possible therapeutic strategies. Finally, we propose that CGG repeat expansion diseases may represent manifestations of a single underlying neuromyodegenerative syndrome in which different organs are affected to different extents depending on the gene location of the repeat expansion., (© 2022. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
266. High-Throughput Methylation-Specific Triplet-Primed PCR and Melting Curve Analysis for Selective and Reliable Identification of Actionable FMR1 Genotypes.
- Author
-
Rajan-Babu IS, Phang GP, Law HY, Lee CG, and Chong SS
- Subjects
- Female, Genotype, Humans, Infant, Newborn, Male, Methylation, Mutation, Polymerase Chain Reaction, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics
- Abstract
Methylated FMR1 full-mutation expansions cause fragile X syndrome. FMR1 premutation carriers are susceptible to other late-onset conditions, and women with premutation are at risk of transmitting a fully expanded FMR1 allele to offspring. Identification of individuals with actionable FMR1 genotypes (full-mutation males and females, and premutation females at risk for primary ovarian insufficiency and/or having fragile X-affected offspring) can enable timely access to intervention services and genetic counseling. This study presents a rapid, first-tier test based on melting curve analysis of methylation-specific triplet-primed PCR amplicons (msTP-PCR MCA) for concurrent detection of FMR1 CGG-repeat expansions and their methylation status. The msTP-PCR MCA assay was optimized on 20 fragile X reference samples, and its performance was evaluated on 111 peripheral blood-derived DNA samples from patients who have undergone prior molecular testing with PCR and/or Southern blot analysis. The msTP-PCR MCA assay detected all samples with a methylated FMR1 CGG-repeat expansion, and had sensitivity, specificity, positive predictive value, and negative predictive values of 100%, 92.06%, 91.1%, and 100%, respectively. The msTP-PCR MCA assay identified premutation/full-mutation mosaicism down to 1%, detected skewed inactivation in females with FMR1 expansions, and enabled selective identification of all individuals with an actionable FMR1 genotype. The msTP-PCR MCA assay may aid in fragile X screening of at-risk populations and newborns and voluntary carrier screening of women of reproductive age., (Copyright © 2022 Association for Molecular Pathology and American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
267. Recurrent missense variant in the nuclear export signal of FMR1 associated with FXS-like phenotype including intellectual disability, ASD, facial abnormalities.
- Author
-
Mangano GD, Fontana A, Salpietro V, Antona V, Mangano GR, and Nardello R
- Subjects
- Female, Humans, Male, Mutation, Missense, Nuclear Export Signals genetics, Phenotype, Autism Spectrum Disorder complications, Autism Spectrum Disorder genetics, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, Intellectual Disability complications, Intellectual Disability genetics
- Abstract
Fragile X syndrome (FXS; MIM 300624) is an X-linked genetic disorder characterized by physical abnormalities associated with intellectual disability and a wide spectrum of neurological and psychiatric impairments. FXS occurs more frequently in males, 1 in 5000 males and 1 in 8000 females accounting for 1-2% of overall intellectual disability (ID). In more than 99% of patients, FXS results from expansions of a CGG triplet repeat (>200 in male) of the FMR1 gene. In the last years an increasing number, albeit still limited, of FXS subjects carrying FMR1 mutations including deletions, splicing errors, missense, and nonsense variants was reported. Nevertheless, the studies concerning the functional consequences of mutations in the FMR1 gene are rare so far and, therefore, we do not have sufficient knowledge regarding the genotype/phenotype correlation. We report a child carrying a hemizygous missense FMR1 (NM_002024.5:c.1325G > A p.Arg442Gln) variant, maternally inherited, associated with facial abnormalities, developmental delay, and social and communication deficits assessed with formal neuropsychological tests. The study contributes to highlighting the clinical differences between the CGG triplet repeat dependent phenotype and FMR1variant dependent phenotype and it also confirms the pathogenicity of the variant being reported for the second time in the literature., (Copyright © 2022. Published by Elsevier Masson SAS.)
- Published
- 2022
- Full Text
- View/download PDF
268. Molecular screening for fragile X syndrome in children with unexplained intellectual disability and/or autistic behaviour.
- Author
-
Stoyanova M, Hachmeriyan M, Levkova M, Bichev S, Georgieva M, Mladenov V, and Angelova L
- Subjects
- Child, Fragile X Mental Retardation Protein genetics, Humans, Autistic Disorder diagnosis, Autistic Disorder genetics, Fragile X Syndrome complications, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Intellectual Disability diagnosis, Intellectual Disability genetics
- Abstract
Introduction: Fragile X syndrome (FXS, OMIM #300624) is the most common inherited form of intellectual disability and the leading monogenic cause of autism., (This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2022
- Full Text
- View/download PDF
269. FXS causing missense mutations disrupt FMRP granule formation, dynamics, and function.
- Author
-
Starke EL, Zius K, and Barbee SA
- Subjects
- Animals, Drosophila genetics, Drosophila metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Mutation, Missense, Neurons metabolism, Fragile X Syndrome genetics
- Abstract
Fragile X Syndrome (FXS) is the most prevalent cause of inherited mental deficiency and is the most common monogenetic cause of autism spectral disorder (ASD). Here, we demonstrate that disease-causing missense mutations in the conserved K homology (KH) RNA binding domains (RBDs) of FMRP cause defects in its ability to form RNA transport granules in neurons. Using molecular, genetic, and imaging approaches in the Drosophila FXS model system, we show that the KH1 and KH2 domains of FMRP regulate distinct aspects of neuronal FMRP granule formation, dynamics, and transport. Furthermore, mutations in the KH domains disrupt translational repression in cells and the localization of known FMRP target mRNAs in neurons. These results suggest that the KH domains play an essential role in neuronal FMRP granule formation and function which may be linked to the molecular pathogenesis of FXS., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
270. FMRP Sustains Presynaptic Function via Control of Activity-Dependent Bulk Endocytosis.
- Author
-
Bonnycastle K, Kind PC, and Cousin MA
- Subjects
- Animals, Endocytosis, Large-Conductance Calcium-Activated Potassium Channels genetics, Large-Conductance Calcium-Activated Potassium Channels metabolism, Male, Neurotransmitter Agents genetics, Neurotransmitter Agents metabolism, Rats, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, Fragile X Syndrome metabolism
- Abstract
Synaptic vesicle (SV) recycling is essential for the maintenance of neurotransmission, with a number of neurodevelopmental disorders linked to defects in this process. Fragile X syndrome (FXS) results from a loss of fragile X mental retardation protein (FMRP) encoded by the FMR1 gene. Hyperexcitability of neuronal circuits is a key feature of FXS, therefore we investigated whether SV recycling was affected by the absence of FMRP during increased neuronal activity. We revealed that primary neuronal cultures from male Fmr1 knock-out (KO) rats display a specific defect in activity-dependent bulk endocytosis (ADBE). ADBE is dominant during intense neuronal activity, and this defect resulted in an inability of Fmr1 KO neurons to sustain SV recycling during trains of high-frequency stimulation. Using a molecular replacement strategy, we also revealed that a human FMRP mutant that cannot bind BK channels failed to correct ADBE dysfunction in KO neurons, however this dysfunction was corrected by BK channel agonists. Therefore, FMRP performs a key role in sustaining neurotransmitter release via selective control of ADBE, suggesting intervention via this endocytosis mode may correct the hyperexcitability observed in FXS. SIGNIFICANCE STATEMENT Loss of fragile X mental retardation protein (FMRP) results in fragile X syndrome (FXS), however whether its loss has a direct role in neurotransmitter release remains a matter of debate. We demonstrate that neurons lacking FMRP display a specific defect in a mechanism that sustains neurotransmitter release during intense neuronal firing, called activity-dependent bulk endocytosis (ADBE). This discovery provides key insights into mechanisms of brain communication that occur because of loss of FMRP function. Importantly it also reveals ADBE as a potential therapeutic target to correct the circuit hyperexcitability observed in FXS., (Copyright © 2022 the authors.)
- Published
- 2022
- Full Text
- View/download PDF
271. Implications of Poly(A) Tail Processing in Repeat Expansion Diseases.
- Author
-
Joachimiak P, Ciesiołka A, Figura G, and Fiszer A
- Subjects
- Humans, Polyadenylation, RNA, Messenger genetics, Fragile X Syndrome genetics, Huntington Disease genetics
- Abstract
Repeat expansion diseases are a group of more than 40 disorders that affect mainly the nervous and/or muscular system and include myotonic dystrophies, Huntington's disease, and fragile X syndrome. The mutation-driven expanded repeat tract occurs in specific genes and is composed of tri- to dodeca-nucleotide-long units. Mutant mRNA is a pathogenic factor or important contributor to the disease and has great potential as a therapeutic target. Although repeat expansion diseases are quite well known, there are limited studies concerning polyadenylation events for implicated transcripts that could have profound effects on transcript stability, localization, and translation efficiency. In this review, we briefly present polyadenylation and alternative polyadenylation (APA) mechanisms and discuss their role in the pathogenesis of selected diseases. We also discuss several methods for poly(A) tail measurement (both transcript-specific and transcriptome-wide analyses) and APA site identification-the further development and use of which may contribute to a better understanding of the correlation between APA events and repeat expansion diseases. Finally, we point out some future perspectives on the research into repeat expansion diseases, as well as APA studies.
- Published
- 2022
- Full Text
- View/download PDF
272. Fragile X Syndrome: From Molecular Aspect to Clinical Treatment.
- Author
-
Protic DD, Aishworiya R, Salcedo-Arellano MJ, Tang SJ, Milisavljevic J, Mitrovic F, Hagerman RJ, and Budimirovic DB
- Subjects
- Animals, Brain pathology, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome pathology, Humans, Phenotype, Fragile X Syndrome genetics
- Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the full mutation as well as highly localized methylation of the fragile X mental retardation 1 ( FMR1 ) gene on the long arm of the X chromosome. Children with FXS are commonly co-diagnosed with Autism Spectrum Disorder, attention and learning problems, anxiety, aggressive behavior and sleep disorder, and early interventions have improved many behavior symptoms associated with FXS. In this review, we performed a literature search of original and review articles data of clinical trials and book chapters using MEDLINE (1990-2021) and ClinicalTrials.gov. While we have reviewed the biological importance of the fragile X mental retardation protein (FMRP), the FXS phenotype, and current diagnosis techniques, the emphasis of this review is on clinical interventions. Early non-pharmacological interventions in combination with pharmacotherapy and targeted treatments aiming to reverse dysregulated brain pathways are the mainstream of treatment in FXS. Overall, early diagnosis and interventions are fundamental to achieve optimal clinical outcomes in FXS.
- Published
- 2022
- Full Text
- View/download PDF
273. Recent research in fragile X-associated tremor/ataxia syndrome.
- Author
-
Salcedo-Arellano MJ and Hagerman RJ
- Subjects
- Ataxia genetics, Ataxia pathology, Female, Fragile X Mental Retardation Protein genetics, Humans, Male, Trinucleotide Repeat Expansion genetics, Fragile X Syndrome genetics, Tremor genetics, Tremor pathology
- Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a cytosine-guanine-guanine repeat expansion neurological disease that occurs in a subset of aging carriers of the premutation (55-200 cytosine-guanine-guanine repeats) in the FMR1 gene located on the X chromosome. The clinical core involves intention tremor and gait ataxia. Current research seeks to clarify the pathophysiology and neuropathology of FXTAS, as well as the development of useful biomarkers to track the progression of FXTAS. Efforts to implement quantitative measures of clinical features, such as kinematics and cognitive measures, are of special interest, in addition to characterize the differences in progression in males compared with females and the efficacy of new treatments., Competing Interests: Conflict of interest statement Nothing declared., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
274. Mitochondrial bioenergetics of astrocytes in Fragile X syndrome: new perspectives on culture conditions and sex effects.
- Author
-
Vandenberg GG, Thotakura A, and Scott AL
- Subjects
- Animals, Cell Hypoxia physiology, Cells, Cultured, Cerebral Cortex metabolism, Female, Fragile X Mental Retardation Protein antagonists & inhibitors, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Male, Mice, Mice, 129 Strain, Mice, Knockout, Mitochondria genetics, Reactive Oxygen Species metabolism, Astrocytes metabolism, Energy Metabolism physiology, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome metabolism, Mitochondria metabolism, Sex Characteristics
- Abstract
Fragile X syndrome (FXS) is a genetic disorder that is characterized by a range of cognitive and behavioral deficits, including mild-moderate intellectual disability. The disease is characterized by an X-linked mutation of the Fmr1 gene, which causes silencing of the gene coding for fragile X mental retardation protein (FMRP), a translational regulator integral for neurodevelopment. Mitochondrial dysfunction has been recently associated with FXS, with reports of increases in oxidative stress markers, reactive oxygen species, and lipid peroxidation being present in the brain tissue. Astrocytes, a prominent glial cell within the central nervous system (CNS), play a large role in regulating oxidative homeostasis within the developing brain and dysregulation of astrocyte redox balance in FXS, which may contribute to oxidative stress. Astrocyte function and mitochondrial bioenergetics are significantly influenced by oxygen availability and circulating sex hormones; yet, these parameters are rarely considered during in vitro experimentation. Given that the brain normally develops in a range of hypoxic conditions and FXS is a sex-linked genetic disorder, we investigated how different oxygen levels (normoxic vs. hypoxic) and biological sex affected mitochondrial bioenergetics of astrocytes in FXS. Our results demonstrate that both mitochondrial respiration capacity and reactive oxygen species emission are altered with Fmr1 deletion in astrocytes and these changes were dependent upon both sexual dimorphism and oxygen availability.
- Published
- 2022
- Full Text
- View/download PDF
275. Maternal Pragmatic Language Difficulties in the FMR1 Premutation and the Broad Autism Phenotype: Associations with Individual and Family Outcomes.
- Author
-
Klusek J, Thurman AJ, and Abbeduto L
- Subjects
- Depression, Family, Female, Fragile X Mental Retardation Protein genetics, Humans, Loneliness, Mothers, Phenotype, Autism Spectrum Disorder genetics, Fragile X Syndrome genetics, Language
- Abstract
Broader phenotypes associated with genetic liability, including mild difficulties with pragmatic language skills, have been documented in mothers of children with autism spectrum disorder (ASD) and mothers of children with fragile X syndrome (FXS). This study investigated the relationship between pragmatic difficulties and indicators of maternal well-being and family functioning. Pragmatic difficulty was associated with loneliness in mothers of children with ASD or FXS, and with depression, decreased life satisfaction, and poorer family relationship quality in mothers of children with FXS only. Results inform subtle maternal pragmatic language difficulties as a risk factor that that may contribute to reduced health and well-being, informing tailored support services to better meet the unique needs of families of children with ASD or FXS., (© 2021. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
276. Behavior Problems and Social Competence in Fragile X Syndrome: A Systematic Review.
- Author
-
Cregenzán-Royo O, Brun-Gasca C, and Fornieles-Deu A
- Subjects
- Child, Humans, Social Skills, Autistic Disorder epidemiology, Fragile X Syndrome epidemiology, Fragile X Syndrome genetics, Intellectual Disability, Problem Behavior
- Abstract
Fragile X syndrome (FXS) causes intellectual disability and is the known leading cause of autism. Common problems in FXS include behavior and social problems. Along with syndromic characteristics and autism comorbidity, environmental factors might influence these difficulties. This systematic review focuses on the last 20 years of studies concerning behavior and social problems in FXS, considering environmental and personal variables that might influence both problems. Three databases were reviewed, leading to fifty-one studies meeting the inclusion criteria. Attention deficit hyperactivity disorder (ADHD) problems remain the greatest behavior problems, with behavioral problems and social competence being stable during the 20 years. Some developmental trajectories might have changed due to higher methodological control, such as aggressive behavior and attention problems. The socialization trajectory from childhood to adolescence remains unclear. Comorbidity with autism in individuals with FXS increased behavior problems and worsened social competence profiles. At the same time, comparisons between individuals with comorbid FXS and autism and individuals with autism might help define the comorbid phenotype. Environmental factors and parental characteristics influenced behavior problems and social competence. Higher methodological control is needed in studies including autism symptomatology and parental characteristics. More studies comparing autism in FXS with idiopathic autism are needed to discern differences between conditions.
- Published
- 2022
- Full Text
- View/download PDF
277. The Fragile X Proteins Differentially Regulate Translation of Reporter mRNAs with G-quadruplex Structures.
- Author
-
Edwards M and Joseph S
- Subjects
- Autism Spectrum Disorder, Binding Sites, Humans, Protein Binding, Protein Domains, RNA metabolism, RNA Stability, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, G-Quadruplexes, RNA, Messenger genetics
- Abstract
Fragile X Syndrome, as well as some manifestations of autism spectrum disorder, results from improper RNA regulation due to a deficiency of fragile X mental retardation protein (FMRP). FMRP and its autosomal paralogs, fragile X related proteins 1 & 2 (FXR1P/2P), have been implicated in many aspects of RNA regulation, from protein synthesis to mRNA stability and decay. The literature on the fragile X related proteins' (FXPs) role in mRNA regulation and their potential mRNA targets is vast. Therefore, we developed an approach to investigate the function of FXPs in translational control using three potential mRNA targets. Briefly, we first selected top mRNA candidates found to be associated with the FXPs and whose translation are influenced by one or more of the FXPs. We then narrowed down the FXPs' binding site(s) within the mRNA, analyzed the strength of this binding in vitro, and determined how each FXP affects the translation of a minimal reporter mRNA with the binding site. Overall, all FXPs bound with high affinity to RNAs containing G-quadruplexes, such as Cyclin Dependent Kinase Inhibitor p21 and FMRP's own coding region. Interestingly, FMRP inhibited the translation of each mRNA distinctly and in a manner that appears to correlate with its binding to each mRNA. In contrast, FXR1P/2P inhibited all mRNAs tested. Finally, although binding of our RNAs was due to the RGG (arginine-glycine-glycine) motif-containing C-terminal region of the FXPs, this region was not sufficient to cause inhibition of translation., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
278. Family history of FXTAS is associated with age-related cognitive-linguistic decline among mothers with the FMR1 premutation.
- Author
-
Klusek J, Fairchild A, Moser C, Mailick MR, Thurman AJ, and Abbeduto L
- Subjects
- Adult, Alleles, Ataxia genetics, Child, Female, Humans, Middle Aged, Mothers, Neurodegenerative Diseases complications, Neurodegenerative Diseases genetics, Tremor genetics, Cognitive Dysfunction complications, Cognitive Dysfunction genetics, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome complications, Fragile X Syndrome genetics, Language Disorders
- Abstract
Background: Women who carry a premutation allele of the FMR1 gene are at increased vulnerability to an array of age-related symptoms and disorders, including age-related decline in select cognitive skills. However, the risk factors for age-related decline are poorly understood, including the potential role of family history and genetic factors. In other forms of pathological aging, early decline in syntactic complexity is observed and predicts the later onset of neurodegenerative disease. To shed light on the earliest signs of degeneration, the present study characterized longitudinal changes in the syntactic complexity of women with the FMR1 premutation across midlife, and associations with family history of fragile X-associated tremor/ataxia syndrome (FXTAS) and CGG repeat length., Methods: Forty-five women with the FMR1 premutation aged 35-64 years at study entry participated in 1-5 longitudinal assessments spaced approximately a year apart (130 observations total). All participants were mothers of children with confirmed fragile X syndrome. Language samples were analyzed for syntactic complexity and participants provided information on family history of FXTAS. CGG repeat length was determined via molecular genetic testing., Results: Hierarchical linear models indicated that women who reported a family history of FXTAS exhibited faster age-related decline in syntactic complexity than those without a family history, with that difference emerging as the women reached their mid-50 s. CGG repeat length was not a significant predictor of age-related change., Conclusions: Results suggest that women with the FMR1 premutation who have a family history of FXTAS may be at increased risk for neurodegenerative disease, as indicated by age-related loss of syntactic complexity. Thus, family history of FXTAS may represent a personalized risk factor for age-related disease. Follow-up study is needed to determine whether syntactic decline is an early indicator of FXTAS specifically, as opposed to being a more general age-related cognitive decline associated with the FMR1 premutation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
279. Fragile X Syndrome: Lessons Learned and What New Treatment Avenues Are on the Horizon.
- Author
-
Hagerman RJ and Hagerman PJ
- Subjects
- Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Mental Retardation Protein therapeutic use, Humans, Autism Spectrum Disorder drug therapy, Autism Spectrum Disorder genetics, Fragile X Syndrome drug therapy, Fragile X Syndrome genetics, Fragile X Syndrome metabolism
- Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading single-gene form of autism spectrum disorder, encompassing cognitive, behavioral, and physical forms of clinical involvement. FXS is caused by large expansions of a noncoding CGG repeat (>200 repeats) in the FMR1 gene, at which point the gene is generally silenced. Absence of FMR1 protein (FMRP), important for synaptic development and maintenance, gives rise to the neurodevelopmental disorder. There is, at present, no therapeutic approach that directly reverses the loss of FMRP; however, there is an increasing number of potential treatments that target the pathways dysregulated in FXS, including those that address the enhanced activity of the mGluR5 pathway and deficits in GABA pathways. Based on studies of targeted therapeutics to date, the prospects are good for one or more effective therapies for FXS in the near future.
- Published
- 2022
- Full Text
- View/download PDF
280. [The cellular functions of G-quadruplex in neurological diseases].
- Author
-
Shioda N
- Subjects
- Guanine, Humans, RNA, Fragile X Syndrome drug therapy, Fragile X Syndrome genetics, G-Quadruplexes, Nervous System Diseases drug therapy, Nervous System Diseases genetics
- Abstract
G-quadruplex (G4) is a unique nucleic acid structure that formed when a four-stranded structure is produced within a single-stranded guanine-rich sequence. Four guanine molecules form a square planar arrangement, termed G-quartet, which are stacked on top of each other to form the G4 structure in DNA (G4DNA) and in RNA (G4RNA). Recent studies have revealed that G4DNA and G4RNA are folded in cells, which suggested their biological and pharmacological significance in DNA replication, transcription, epigenetic modification, and RNA metabolism. So far, we have reported the following; 1) G4 is a target of cognitive function therapy for ATR-X intellectual disability syndrome, in which mutations are found in a G4 binding protein ATRX. 2) G4 is formed in heterochromatin depending on neuronal development. 3) G4 promotes prionoids in a CGG triplet repeat disease, Fragile X-associated tremor/ataxia syndrome (FXTAS). 4) 5-aminolevulinic acid is a potential candidate drug for treating some neurological diseases through the G4 binding ability. In this review, we summarized the significant roles of G4 in neurological diseases.
- Published
- 2022
- Full Text
- View/download PDF
281. Paternal retraction of a fragile X allele to normal size, showing normal function over two generations.
- Author
-
Bartlett E, Archibald AD, Francis D, Ling L, Thomas R, Chandler G, Ward L, O'Farrell G, Pandelache A, Delatycki MB, Bennetts BH, Ho G, Fisk K, Baker EK, Amor DJ, and Godler DE
- Subjects
- Alleles, DNA Methylation, Female, Humans, Male, Mutation, Trinucleotide Repeat Expansion, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics
- Abstract
The FMR1 premutation (PM:55-199 CGG) is associated with fragile X-associated tremor/ataxia syndrome (FXTAS) and when maternally transmitted is at risk of expansion to a hypermethylated full mutation (FM: ≥ 200 CGG) that causes fragile X syndrome (FXS). We describe a maternally transmitted PM (77 CGG) that was passed to a son (103 CGG), and to a daughter (220-1822 CGG), who were affected with FXTAS and FXS, respectively. The male with the PM showed low-level mosaicism for normal size of 30 and 37 CGG. This male had two offspring: one female mosaic for PM and FM (56, 157, >200 CGG) and another with only a 37 CGG allele detected in multiple tissues, neither with a clinical phenotype. The female with the 37 CGG allele showed normal levels of FMR1 methylation and mRNA and passed this 37 CGG allele to one of her daughters, who was also unaffected. These findings show that post-zygotic paternal retraction can lead to low-level mosaicism for normal size alleles, with these normal alleles being functional when passed over two generations., (© 2021 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
282. [Monogenetic causes of psychiatric disorders: a review].
- Author
-
van Amelsvoort TAMJ and Swillen A
- Subjects
- Comorbidity, Humans, Psychopathology, Fragile X Syndrome genetics, Mental Disorders epidemiology, Mental Disorders genetics, Psychiatry
- Abstract
Background: Because of rapid developments in genetic technology, more underlying genetic causes of psychiatric disorders can be detected which may contribute to better monitoring and treatment of co-morbidities than previously., Aim: Review of monogenetic causes of psychiatric disorders., Methode: Review of the literature., Resultats: Research in people with monogenetic disorders will generate new knowledge and insights on psychopathology and cognitive function in general and pave the way to new treatment targets. In this article we discuss four monogenetic disorders that are relevant for clinical psychiatry and (educational) psychology: fragile X syndrome, tuberous sclerosis, Rett Syndrome, and Huntington’s disease., Conclusion: Given the multisystem nature of these genetic disorders, a well-coordinated, multidisciplinary approach by specialized expert centers is highly recommended.
- Published
- 2022
283. Functional consequences of postnatal interventions in a mouse model of Fragile X syndrome.
- Author
-
Rais M, Lovelace JW, Shuai XS, Woodard W, Bishay S, Estrada L, Sharma AR, Nguy A, Kulinich A, Pirbhoy PS, Palacios AR, Nelson DL, Razak KA, and Ethell IM
- Subjects
- Animals, Disease Models, Animal, Fragile X Mental Retardation Protein genetics, Mice, Mice, Knockout, Neurons physiology, Fragile X Syndrome genetics
- Abstract
Background: Fragile X syndrome (FXS) is a leading genetic cause of autism and intellectual disability with cortical hyperexcitability and sensory hypersensitivity attributed to loss and hypofunction of inhibitory parvalbumin-expressing (PV) cells. Our studies provide novel insights into the role of excitatory neurons in abnormal development of PV cells during a postnatal period of inhibitory circuit refinement., Methods: To achieve Fragile X mental retardation gene (Fmr1) deletion and re-expression in excitatory neurons during the postnatal day (P)14-P21 period, we generated Cre
CaMKIIa /Fmr1Flox/y (cOFF) and CreCaMKIIa /Fmr1FloxNeo/y (cON) mice, respectively. Cortical phenotypes were evaluated in adult mice using biochemical, cellular, clinically relevant electroencephalogram (EEG) and behavioral tests., Results: We found that similar to global Fmr1 KO mice, the density of PV-expressing cells, their activation, and sound-evoked gamma synchronization were impaired in cOFF mice, but the phenotypes were improved in cON mice. cOFF mice also showed enhanced cortical gelatinase activity and baseline EEG gamma power, which were reduced in cON mice. In addition, TrkB phosphorylation and PV levels were lower in cOFF mice, which also showed increased locomotor activity and anxiety-like behaviors. Remarkably, when FMRP levels were restored in only excitatory neurons during the P14-P21 period, TrkB phosphorylation and mouse behaviors were also improved., Conclusions: These results indicate that postnatal deletion or re-expression of FMRP in excitatory neurons is sufficient to elicit or ameliorate structural and functional cortical deficits, and abnormal behaviors in mice, informing future studies about appropriate treatment windows and providing fundamental insights into the cellular mechanisms of cortical circuit dysfunction in FXS., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
284. Multiple Early-Life Seizures Alters Neonatal Communicative Behavior in Fmr1 Knockout Mice.
- Author
-
Nguyen PH, Narvaiz DA, Womble PD, Sullens DG, Binder MS, Hodges SL, Kwok E, and Lugo JN
- Subjects
- Animals, Male, Mice, Vocalization, Animal, Mice, Knockout, Mice, Inbred C57BL, Fragile X Mental Retardation Protein genetics, Seizures, Disease Models, Animal, Autism Spectrum Disorder, Fragile X Syndrome complications, Fragile X Syndrome genetics
- Abstract
Fragile X syndrome (FXS) is the leading monogenic cause of intellectual disability and a significant contributor to Autism Spectrum Disorder. Individuals with FXS are subject to developing numerous comorbidities, one of the most prevalent being seizures. In the present study, we investigated how seizures affected neonatal communicative behavior in the FXS mouse model. On postnatal day (PD) 7 through 11, we administered 3 flurothyl seizures per day to both Fmr1 knockout and wild-type C57BL/6J male mice. Ultrasonic vocalizations were recorded on PD12. Statistically significant alterations were found in both spectral and temporal measurements across seizure groups. We found that induction of seizures across PD7-11 resulted in an increased fundamental frequency (pitch) of ultrasonic vocalizations produced (p < 0.05), a longer duration of calls (p < 0.05), and a greater cumulative duration of calls (p < 0.05) in both genotypes. Induction of seizures across PD7-11 also resulted in a decreased latency to the first emitted vocalization (p < 0.05) and a decrease in mean power (loudness) for their vocalizations (p < 0.05). Early-life seizures also resulted in an increase in the number of downward and frequency step call types (p < 0.05). There was a significant increase in the number of chevron calls emitted from the Fmr1 knockout mice that received seizures compared to knockout control and wild-type seizure mice (p < 0.05). Overall, this study provides evidence that early-life seizures result in communication impairments and that superimposing seizures in Fmr1 knockout mice does produce an additional deficit in vocalization., (© 2022 S. Karger AG, Basel.)
- Published
- 2022
- Full Text
- View/download PDF
285. Raising Knowledge and Awareness of Fragile X Syndrome in Serbia, Georgia, and Colombia: A Model for Other Developing Countries?
- Author
-
Protic D, Salcedo-Arellano MJ, Stojkovic M, Saldarriaga W, Ávila Vidal LA, Miller RM, Tabatadze N, Peric M, Hagerman R, and Budimirovic DB
- Subjects
- Autism Spectrum Disorder, Colombia epidemiology, Cross-Sectional Studies, Developing Countries, Fragile X Mental Retardation Protein, Georgia (Republic) epidemiology, Humans, Mutation, Serbia epidemiology, Fragile X Syndrome epidemiology, Fragile X Syndrome genetics
- Abstract
Fragile X syndrome is the most common monogenetic cause of inherited intellectual disability and syndromic autism spectrum disorder. Fragile X syndrome is caused by an expansion (full mutation ≥200 CGGs repeats, normal 10-45 CGGs) of the fragile X mental retardation 1 ( FMR1 ) gene, epigenetic silencing of the gene, which leads to reduction or lack of the gene's product: the fragile X mental retardation protein. In this cross-sectional study, we assessed general and pharmacotherapy knowledge (GK and PTK) of fragile X syndrome and satisfaction with education in neurodevelopmental disorders (NDDs) among senior medical students in Serbia (N=348), Georgia (N=112), and Colombia (N=58). A self-administered 18-item questionnaire included GK (8/18) and PTK (7/18) components and self-assessment of the participants education in NDDs (3/18). Roughly 1 in 5 respondents had correct answers on half or more facts about fragile X syndrome (GK>PTK), which ranged similarly 5-7 in Serbia, 6-8 in Georgia, and 5-8 in Colombia, respectively. No cohort had an average value greater than 9 (60%) that would represent passing score "cut-off." None of the participants answered all the questions correctly. More than two-thirds of the participants concluded that they gained inadequate knowledge of NDDs during their studies, and that their education in this field should be more intense. In conclusion, there is a major gap in knowledge regarding fragile X syndrome among senior medical students in these three developing countries. The finding could at least in part be generalized to other developing countries aimed toward increasing knowledge and awareness of NDDs and fostering an institutional collaboration between developed and developing countries., (Copyright ©2021, Yale Journal of Biology and Medicine.)
- Published
- 2021
286. Screening for FMR1 CGG Repeat Expansion in Thai Patients with Autism Spectrum Disorder.
- Author
-
Hnoonual A, Jankittunpaiboon C, and Limprasert P
- Subjects
- Alleles, Autistic Disorder genetics, Child, Preschool, Female, Fragile X Syndrome genetics, Humans, Male, Mass Screening methods, Mutation genetics, Pilot Projects, Thailand, Autism Spectrum Disorder genetics, Fragile X Mental Retardation Protein genetics
- Abstract
Autism spectrum disorder (ASD) is a complex disorder with a heterogeneous etiology. Fragile X syndrome (FXS) is recognized as the most common single gene mutation associated with ASD. FXS patients show some autistic behaviors and may be difficult to distinguish at a young age from autistic children. However, there have been no published reports on the prevalence of FXS in ASD patients in Thailand. In this study, we present a pilot study to analyze the CGG repeat sizes of the FMR1 gene in Thai autistic patients. We screened 202 unrelated Thai patients (168 males and 34 females) with nonsyndromic ASD and 212 normal controls using standard FXS molecular diagnosis techniques. The distributions of FMR1 CGG repeat sizes in the ASD and normal control groups were similar, with the two most common alleles having 29 and 30 CGG repeats, followed by an allele with 36 CGG repeats. No FMR1 full mutations or premutations were found in either ASD individuals or the normal controls. Interestingly, three ASD male patients with high normal CGG and intermediate CGG repeats (44, 46, and 53 CGG repeats) were identified, indicating that the prevalence of FMR1 intermediate alleles in Thai ASD patients was approximately 1% while these alleles were absent in the normal male controls. Our study indicates that CGG repeat expansions of the FMR1 gene may not be a common genetic cause of nonsyndromic ASD in Thai patients. However, further studies for mutations other than the CGG expansion in the FMR1 gene are required to get a better information on FXS prevalence in Thai ASD patients., Competing Interests: All authors declare that they have no conflicts of interest., (Copyright © 2021 Areerat Hnoonual et al.)
- Published
- 2021
- Full Text
- View/download PDF
287. Urokinase plasminogen activator mediates changes in human astrocytes modeling fragile X syndrome.
- Author
-
Peteri UK, Pitkonen J, de Toma I, Nieminen O, Utami KH, Strandin TM, Corcoran P, Roybon L, Vaheri A, Ethell I, Casarotto P, Pouladi MA, and Castrén ML
- Subjects
- Animals, Astrocytes metabolism, Humans, Mice, Urokinase-Type Plasminogen Activator metabolism, Autism Spectrum Disorder metabolism, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Induced Pluripotent Stem Cells metabolism
- Abstract
The function of astrocytes intertwines with the extracellular matrix, whose neuron and glial cell-derived components shape neuronal plasticity. Astrocyte abnormalities have been reported in the brain of the mouse model for fragile X syndrome (FXS), the most common cause of inherited intellectual disability, and a monogenic cause of autism spectrum disorder. We compared human FXS and control astrocytes generated from human induced pluripotent stem cells and we found increased expression of urokinase plasminogen activator (uPA), which modulates degradation of extracellular matrix. Several pathways associated with uPA and its receptor function were activated in FXS astrocytes. Levels of uPA were also increased in conditioned medium collected from FXS hiPSC-derived astrocyte cultures and correlated inversely with intracellular Ca
2+ responses to activation of L-type voltage-gated calcium channels in human astrocytes. Increased uPA augmented neuronal phosphorylation of TrkB within the docking site for the phospholipase-Cγ1 (PLCγ1), indicating effects of uPA on neuronal plasticity. Gene expression changes during neuronal differentiation preceding astrogenesis likely contributed to properties of astrocytes with FXS-specific alterations that showed specificity by not affecting differentiation of adenosine triphosphate (ATP)-responsive astrocyte population. To conclude, our studies identified uPA as an important regulator of astrocyte function and demonstrated that increased uPA in human FXS astrocytes modulated astrocytic responses and neuronal plasticity., (© 2021 The Authors. GLIA published by Wiley Periodicals LLC.)- Published
- 2021
- Full Text
- View/download PDF
288. FMR1 allele frequencies in 51,000 newborns: a large-scale population study in China.
- Author
-
Zhang JY, Wu DW, Yang RL, Zhu L, Jiang MY, Wang WJ, Li XK, Jiang XL, Tong F, and Shu Q
- Subjects
- Alleles, Female, Gene Frequency, Humans, Infant, Newborn, Male, Mutation, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome epidemiology, Fragile X Syndrome genetics
- Abstract
Background: Fragile X syndrome (FXS), caused by CGG-repeat expansion in FMR1 promoter, is one of the most common causes of mental retardation. Individuals with full mutation and premutation alleles have a high risk of psychophysiological disorder and of having affected offspring. Frequencies of FMR1 alleles in general newborns have been reported in Caucasians but have not been investigated in the large-scale population in the mainland of China., Methods: The sizes of FMR1 CGG-repeats were analyzed in 51,661 newborns (28,114 males and 23,547 females) and also in a cohort of 33 children diagnosed with developmental delay using GC-rich polymerase chain reaction (PCR) and triple repeat primed PCR., Results: The frequency of CGG repeats > 100 was 1/9371 in males and 1/5887 in females, and the frequency of CGG repeats > 54 was 1/1561 in males and 1/1624 in females. FMR1 full mutation and premutation were identified in 27.27% of children who had Ages and Stages Questionnaire scores less than two standard deviations from the cutoff value., Conclusions: Our study revealed the prevalence of FXS in China and improved the sample databases of FXS, suggesting that the prevalence of FXS in Chinese is higher than estimated previously and that FXS screening can be advised to high-risk families., (© 2021. Children's Hospital, Zhejiang University School of Medicine.)
- Published
- 2021
- Full Text
- View/download PDF
289. Auditory hypersensitivity and processing deficits in a rat model of fragile X syndrome.
- Author
-
Auerbach BD, Manohar S, Radziwon K, and Salvi R
- Subjects
- Animals, Autism Spectrum Disorder genetics, Disease Models, Animal, Fragile X Mental Retardation Protein genetics, Mice, Knockout, Rats, Rats, Transgenic, Mice, Fragile X Syndrome complications, Fragile X Syndrome genetics, Hyperacusis
- Abstract
Fragile X (FX) syndrome is one of the leading inherited causes of autism spectrum disorder (ASD). A majority of FX and ASD patients exhibit sensory hypersensitivity, including auditory hypersensitivity or hyperacusis, a condition in which everyday sounds are perceived as much louder than normal. Auditory processing deficits in FX and ASD also afford the opportunity to develop objective and quantifiable outcome measures that are likely to translate between humans and animal models due to the well-conserved nature of the auditory system and well-developed behavioral read-outs of sound perception. Therefore, in this study we characterized auditory hypersensitivity in a Fmr1 knockout (KO) transgenic rat model of FX using an operant conditioning task to assess sound detection thresholds and suprathreshold auditory reaction time-intensity (RT-I) functions, a reliable psychoacoustic measure of loudness growth, at a variety of stimulus frequencies, bandwidths, and durations. Male Fmr1 KO and littermate WT rats both learned the task at the same rate and exhibited normal hearing thresholds. However, Fmr1 KO rats had faster auditory RTs over a broad range of intensities and steeper RT-I slopes than WT controls, perceptual evidence of excessive loudness growth in Fmr1 KO rats. Furthermore, we found that Fmr1 KO animals exhibited abnormal perceptual integration of sound duration and bandwidth, with diminished temporal but enhanced spectral integration of sound intensity. Because temporal and spectral integration of sound stimuli were altered in opposite directions in Fmr1 KO rats, this suggests that abnormal RTs in these animals are evidence of aberrant auditory processing rather than generalized hyperactivity or altered motor responses. Together, these results are indicative of fundamental changes to low-level auditory processing in Fmr1 KO animals. Finally, we demonstrated that antagonism of metabotropic glutamate receptor 5 (mGlu5) selectively and dose-dependently restored normal loudness growth in Fmr1 KO rats, suggesting a pharmacologic approach for alleviating sensory hypersensitivity associated with FX. This study leverages the tractable nature of the auditory system and the unique behavioral advantages of rats to provide important insights into the nature of a centrally important yet understudied aspect of FX and ASD., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
290. Explanatory models for the cause of Fragile X Syndrome in rural Cameroon.
- Author
-
Kengne Kamga K, De Vries J, Nguefack S, Munung NS, and Wonkam A
- Subjects
- Cameroon, Female, Fragile X Mental Retardation Protein genetics, Genetic Counseling, Humans, Male, Fragile X Syndrome genetics, Fragile X Syndrome psychology, Intellectual Disability genetics
- Abstract
Among the myriad causes of intellectual disability (ID), Fragile X Syndrome (FXS) is the leading genetic cause. Yet, little is known of how people affected by this condition make sense of it. The present study aimed to investigate the explanatory models for the causes of FXS in an extended family mainly affected by this condition and members of the village from which they originated in Cameroon. Using an ethnographic approach, 92 participants were interviewed (59 females and 33 males) through 10 focus group discussions and 23 in-depth interviews between April 2018 and February 2020. Data analysis revealed four explanatory models regarding the etiologies of FXS in the community. Firstly, the curse model described a curse from the chief because of the belief that his wives did not mourn his intellectually disabled servant. Secondly, the spiritual model relates FXS to a punishment from God. Thirdly, the socioeconomic model attributes FXS to events in the prenatal and perinatal periods. Finally, the genetic model describes the pattern of inheritance of the disease in the family. This paper helps to understand the explanatory disease models that exist for FXS in rural Cameroon and could inform genetic counseling practices, community genetic education, and policymakers when drafting protocols for public engagement activities., (© 2021 The Authors. Journal of Genetic Counseling published by Wiley Periodicals LLC on behalf of National Society of Genetic Counselors.)
- Published
- 2021
- Full Text
- View/download PDF
291. Eye Movements in Fragile X-Associated Tremor/Ataxia Syndrome.
- Author
-
Hall DA, Leehey MA, Hagerman RJ, and Pelak VS
- Subjects
- Ataxia complications, Ataxia diagnosis, Ataxia genetics, Case-Control Studies, Eye Movements, Female, Fragile X Mental Retardation Protein genetics, Humans, Male, Middle Aged, Fragile X Syndrome complications, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Tremor diagnosis, Tremor genetics
- Abstract
Background: Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder characterized by ataxia, tremor, and parkinsonism. Eye motility abnormalities on the clinical examination of FXTAS patients have not been formally studied., Methods: A case-control study with fragile X gene mutation carriers with and without FXTAS and normal controls was conducted and included a videotaping of ocular items of the International Cooperative Ataxia Rating Scale (ICARS). A neuro-ophthalmologist blinded to gene status rated nystagmus, ocular pursuit, and saccades., Results: Forty-four cases and controls were recruited, with an average age of 55.2 years (±7.4) and 57% women. Gaze-evoked nystagmus was increased in fragile X gene carriers (odds ratio 1.44, 95% confidence interval: 0.33-7.36) but was not statistically significant. There was no difference in ocular pursuit nor saccade dysmetria between cases and controls., Conclusion: The results show that clinical examination findings of ocular abnormalities, using the ICARS oculomotor disorders movement subscale, are not more common in FXTAS or FMR1 premutation carriers than normal controls on examination in the clinic. Examining a larger cohort of patients with FXTAS would be an ideal next step., Competing Interests: The authors report no conflicts of interest., (Copyright © 2020 by North American Neuro-Ophthalmology Society.)
- Published
- 2021
- Full Text
- View/download PDF
292. Population-based carrier screening and prenatal diagnosis of fragile X syndrome in East Asian populations.
- Author
-
Guo Q, Chang YY, Huang CH, Hsiao YS, Hsiao YC, Chiu IF, Zhou Y, Zhang H, and Ko TM
- Subjects
- Adult, Alleles, Female, Fragile X Mental Retardation Protein genetics, Humans, Mutation, Pregnancy, Prenatal Diagnosis, Trinucleotide Repeats, Fragile X Syndrome diagnosis, Fragile X Syndrome epidemiology, Fragile X Syndrome genetics
- Abstract
Identification of carriers of fragile X syndrome (FXS) with the subsequent prenatal diagnosis and knowledge of FXS-associated genetic profiles are essential for intervention in specific populations. We report the results of carrier screening of 39,458 East Asian adult women and prenatal diagnosis from 87 FXS carriers. The prevalence of FXS carriers and full mutation fetuses was estimated to be 1/581 and 1/3124 in East Asian populations, respectively. We confirmed the validity of the current threshold of CGG trinucleotide repeats for FMR1 categorization; the integral risks of full mutation expansion were approximately 6.0%, 43.8%, and 100% for premutation alleles with 55-74, 75-89, and ≥ 90 CGG repeats, respectively. The protective effect of AGG (adenine-guanine-guanine nucleotides) interruption in East Asian populations was validated, which is important in protecting premutation alleles with 75-89 CGG repeats from full mutation expansion. Finally, family history was shown not an effective indicator for FXS carrier screening in East Asian populations, and population-based screening was more cost-effective. This study provides an insight into the largest carrier screening and prenatal diagnosis for FXS in East Asian populations to date. The FXS-associated genetic profiles of East Asian populations are delineated, and population-based carrier screening is shown to be promising for FXS intervention., Competing Interests: Conflict of interest None., (Copyright © 2021 Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, and Genetics Society of China. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
293. Prevalence of Underdiagnosed Fragile X Syndrome in 2 Health Systems.
- Author
-
Movaghar A, Page D, Brilliant M, and Mailick M
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Data Mining, Electronic Health Records, Female, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Genetic Testing, Humans, Infant, Infant, Newborn, Male, Middle Aged, Prevalence, Wisconsin epidemiology, Young Adult, Fragile X Syndrome epidemiology
- Published
- 2021
- Full Text
- View/download PDF
294. FMRP and MOV10 regulate Dicer1 expression and dendrite development.
- Author
-
Lannom MC, Nielsen J, Nawaz A, Shilikbay T, and Ceman S
- Subjects
- Animals, Argonaute Proteins, DEAD-box RNA Helicases genetics, Dendrites metabolism, Female, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Male, Mice, Mice, Inbred C57BL, MicroRNAs, Neuroblastoma, Neuronal Outgrowth, Neurons, RNA Helicases genetics, RNA, Messenger, Ribonuclease III genetics, DEAD-box RNA Helicases metabolism, Fragile X Mental Retardation Protein metabolism, RNA Helicases metabolism, Ribonuclease III metabolism
- Abstract
Fragile X syndrome results from the loss of expression of the Fragile X Mental Retardation Protein (FMRP). FMRP and RNA helicase Moloney Leukemia virus 10 (MOV10) are important Argonaute (AGO) cofactors for miRNA-mediated translation regulation. We previously showed that MOV10 functionally associates with FMRP. Here we quantify the effect of reduced MOV10 and FMRP expression on dendritic morphology. Murine neurons with reduced MOV10 and FMRP phenocopied Dicer1 KO neurons which exhibit impaired dendritic maturation Hong J (2013), leading us to hypothesize that MOV10 and FMRP regulate DICER expression. In cells and tissues expressing reduced MOV10 or no FMRP, DICER expression was significantly reduced. Moreover, the Dicer1 mRNA is a Cross-Linking Immunoprecipitation (CLIP) target of FMRP Darnell JC (2011), MOV10 Skariah G (2017) and AGO2 Kenny PJ (2020). MOV10 and FMRP modulate expression of DICER1 mRNA through its 3'untranslated region (UTR) and introduction of a DICER1 transgene restores normal neurite outgrowth in the Mov10 KO neuroblastoma Neuro2A cell line and branching in MOV10 heterozygote neurons. Moreover, we observe a global reduction in AGO2-associated microRNAs isolated from Fmr1 KO brain. We conclude that the MOV10-FMRP-AGO2 complex regulates DICER expression, revealing a novel mechanism for regulation of miRNA production required for normal neuronal morphology., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
295. Co-Occurrence of Fragile X Syndrome with a Second Genetic Condition: Three Independent Cases of Double Diagnosis.
- Author
-
Tabolacci E, Pomponi MG, Remondini L, Pietrobono R, Orteschi D, Nobile V, Pucci C, Musto E, Pane M, Mercuri EM, Neri G, Genuardi M, Chiurazzi P, and Zollino M
- Subjects
- Adult, Child, Child, Preschool, Female, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Humans, Male, Megalencephaly genetics, Megalencephaly metabolism, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne metabolism, Exome Sequencing methods, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome pathology, Megalencephaly pathology, Muscular Dystrophy, Duchenne pathology, Mutation, Nerve Tissue Proteins genetics, Protein Phosphatase 2 genetics, Transcription Factors genetics
- Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and autism caused by the instability of a CGG trinucleotide repeat in exon 1 of the FMR1 gene. The co-occurrence of FXS with other genetic disorders has only been occasionally reported. Here, we describe three independent cases of FXS co-segregation with three different genetic conditions, consisting of Duchenne muscular dystrophy (DMD), PPP2R5D --related neurodevelopmental disorder, and 2p25.3 deletion. The co-occurrence of DMD and FXS has been reported only once in a young boy, while in an independent family two affected boys were described, the elder diagnosed with FXS and the younger with DMD. This represents the second case in which both conditions coexist in a 5-year-old boy, inherited from his heterozygous mother. The next double diagnosis had never been reported before: through exome sequencing, a girl with FXS who was of 7 years of age with macrocephaly and severe psychomotor delay was found to carry a de novo variant in the PPP2R5D gene. Finally, a maternally inherited 2p25.3 deletion associated with a decreased level of the MYT1L transcript, only in the patient, was observed in a 33-year-old FXS male with severe seizures compared to his mother and two sex- and age-matched controls. All of these patients represent very rare instances of genetic conditions with clinical features that can be modified by FXS and vice versa .
- Published
- 2021
- Full Text
- View/download PDF
296. Dysregulation of GABAA Receptor-Mediated Neurotransmission during the Auditory Cortex Critical Period in the Fragile X Syndrome Mouse Model.
- Author
-
Song YJ, Xing B, Barbour AJ, Zhou C, and Jensen FE
- Subjects
- Animals, Disease Models, Animal, Fragile X Mental Retardation Protein genetics, Mice, Mice, Knockout, Receptors, GABA-A, Synaptic Transmission, Auditory Cortex, Fragile X Syndrome genetics
- Abstract
Fragile X syndrome (FXS) is the leading monogenic form of intellectual disability and autism, with patients exhibiting numerous auditory-related phenotypes during their developmental period, including communication, language development, and auditory processing deficits. Despite FXS studies describing excitatory-inhibitory (E-I) imbalance in the auditory circuit and an impaired auditory critical period, evaluation of E-I circuitry maturation in the auditory cortex of FXS models remains limited. Here, we examined GABAA receptor (GABAAR)-mediated inhibitory synaptic transmission within the auditory cortex, characterizing normal intracortical circuit development patterns in wild-type (WT) mice and examining their dysregulation in developing Fmr1 knock-out (KO) mice. Electrophysiological recordings revealed gradual developmental shifts in WT L4-L2/3 connectivity, where circuit excitability significantly increased after critical period onset. KO mice exhibited accelerated developmental shifts related to aberrant GABAergic signaling. Specifically, Fmr1 KO L2/3 pyramidal neurons have enhanced developmental sensitivity to pharmacological GABAAR modulators, altered maturation of GABAAR voltage-dependent conductance, with additional presynaptic GABA release alterations. These differences are further accompanied by alterations in developmental long-term potentiation. Together, our results suggest that altered GABAergic signaling within developing Fmr1 KOs impairs the normal patterning of E-I circuit and synaptic plasticity maturation to contribute to the impaired auditory cortex critical period and functional auditory deficits in FXS., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
297. NMD abnormalities during brain development in the Fmr1-knockout mouse model of fragile X syndrome.
- Author
-
Kurosaki T, Sakano H, Pröschel C, Wheeler J, Hewko A, and Maquat LE
- Subjects
- Animals, Cerebral Cortex metabolism, Disease Models, Animal, Hippocampus metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Neurons metabolism, Brain growth & development, Brain Diseases genetics, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Nonsense Mediated mRNA Decay genetics
- Abstract
Background: Fragile X syndrome (FXS) is an intellectual disability attributable to loss of fragile X protein (FMRP). We previously demonstrated that FMRP binds mRNAs targeted for nonsense-mediated mRNA decay (NMD) and that FMRP loss results in hyperactivated NMD and inhibition of neuronal differentiation in human stem cells., Results: We show here that NMD is hyperactivated during the development of the cerebral cortex, hippocampus, and cerebellum in the Fmr1-knockout (KO) mouse during embryonic and early postnatal periods. Our findings demonstrate that NMD regulates many neuronal mRNAs that are important for mouse brain development., Conclusions: We reveal the abnormal regulation of these mRNAs in the Fmr1-KO mouse, a model of FXS, and highlight the importance of early intervention., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
298. The organization and development of cortical interneuron presynaptic circuits are area specific.
- Author
-
Pouchelon G, Dwivedi D, Bollmann Y, Agba CK, Xu Q, Mirow AMC, Kim S, Qiu Y, Sevier E, Ritola KD, Cossart R, and Fishell G
- Subjects
- Animals, Cerebral Cortex metabolism, Female, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Interneurons metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neural Pathways, Presynaptic Terminals metabolism, Rabies virus genetics, Sense Organs metabolism, Synapses metabolism, Cerebral Cortex pathology, Fragile X Mental Retardation Protein physiology, Fragile X Syndrome pathology, Interneurons pathology, Presynaptic Terminals pathology, Sense Organs pathology, Synapses pathology
- Abstract
Parvalbumin and somatostatin inhibitory interneurons gate information flow in discrete cortical areas that compute sensory and cognitive functions. Despite the considerable differences between areas, individual interneuron subtypes are genetically invariant and are thought to form canonical circuits regardless of which area they are embedded in. Here, we investigate whether this is achieved through selective and systematic variations in their afferent connectivity during development. To this end, we examined the development of their inputs within distinct cortical areas. We find that interneuron afferents show little evidence of being globally stereotyped. Rather, each subtype displays characteristic regional connectivity and distinct developmental dynamics by which this connectivity is achieved. Moreover, afferents dynamically regulated during development are disrupted by early sensory deprivation and in a model of fragile X syndrome. These data provide a comprehensive map of interneuron afferents across cortical areas and reveal the logic by which these circuits are established during development., Competing Interests: Declaration of interests Gord Fishell is a founder of Regel Therapeutics, which has no competing interests with the present manuscript., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
299. SRSF protein kinase 1 modulates RAN translation and suppresses CGG repeat toxicity.
- Author
-
Malik I, Tseng YJ, Wright SE, Zheng K, Ramaiyer P, Green KM, and Todd PK
- Subjects
- Animals, C9orf72 Protein genetics, C9orf72 Protein metabolism, Drosophila melanogaster genetics, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Trinucleotide Repeat Expansion, Amyotrophic Lateral Sclerosis genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism, Fragile X Syndrome genetics, Frontotemporal Dementia, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism
- Abstract
Transcribed CGG repeat expansions cause neurodegeneration in Fragile X-associated tremor/ataxia syndrome (FXTAS). CGG repeat RNAs sequester RNA-binding proteins (RBPs) into nuclear foci and undergo repeat-associated non-AUG (RAN) translation into toxic peptides. To identify proteins involved in these processes, we employed a CGG repeat RNA-tagging system to capture repeat-associated RBPs by mass spectrometry in mammalian cells. We identified several SR (serine/arginine-rich) proteins that interact selectively with CGG repeats basally and under cellular stress. These proteins modify toxicity in a Drosophila model of FXTAS. Pharmacologic inhibition of serine/arginine protein kinases (SRPKs), which alter SRSF protein phosphorylation, localization, and activity, directly inhibits RAN translation of CGG and GGGGCC repeats (associated with C9orf72 ALS/FTD) and triggers repeat RNA retention in the nucleus. Lowering SRPK expression suppressed toxicity in both FXTAS and C9orf72 ALS/FTD model flies, and SRPK inhibitors suppressed CGG repeat toxicity in rodent neurons. Together, these findings demonstrate roles for CGG repeat RNA binding proteins in RAN translation and repeat toxicity and support further evaluation of SRPK inhibitors in modulating RAN translation associated with repeat expansion disorders., (© 2021 The Authors Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
300. The FMR1 Premutation Phenotype and Mother-Youth Synchrony in Fragile X Syndrome.
- Author
-
Moser C, Mattie L, Abbeduto L, and Klusek J
- Subjects
- Adolescent, Executive Function, Female, Fragile X Mental Retardation Protein genetics, Humans, Phenotype, Fragile X Syndrome genetics, Mothers
- Abstract
A subset of mothers who carry the FMR1 premutation may express a unique phenotype. The relationship between the FMR1 phenotype and mother-child interaction in families with fragile X-associated disorders has not been well characterized, despite the importance of high-quality mother-child interaction for child development. This study examined the association between the FMR1 phenotype and the quality of interactions between mothers and their adolescent/young adult sons with fragile X syndrome. Mother-youth synchrony was coded from a dyadic interaction. Maternal anxiety and depression symptoms, executive function deficits, and pragmatic language difficulties were evaluated. Results indicated that pragmatic language was associated with mother-youth synchrony. These findings highlight the importance of family-centered intervention practices for families with fragile X-associated disorders., (©AAIDD.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.