1
|
Maestri S, Scalzo D, Damaggio G, Zobel M, Besusso D, Cattaneo E. Navigating triplet repeats sequencing: concepts, methodological challenges and perspective for Huntington's disease. Nucleic Acids Res 2025; 53:gkae1155. [PMID: 39676657 PMCID: PMC11724279 DOI: 10.1093/nar/gkae1155] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/16/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
The accurate characterization of triplet repeats, especially the overrepresented CAG repeats, is increasingly relevant for several reasons. First, germline expansion of CAG repeats above a gene-specific threshold causes multiple neurodegenerative disorders; for instance, Huntington's disease (HD) is triggered by >36 CAG repeats in the huntingtin (HTT) gene. Second, extreme expansions up to 800 CAG repeats have been found in specific cell types affected by the disease. Third, synonymous single nucleotide variants within the CAG repeat stretch influence the age of disease onset. Thus, new sequencing-based protocols that profile both the length and the exact nucleotide sequence of triplet repeats are crucial. Various strategies to enrich the target gene over the background, along with sequencing platforms and bioinformatic pipelines, are under development. This review discusses the concepts, challenges, and methodological opportunities for analyzing triplet repeats, using HD as a case study. Starting with traditional approaches, we will explore how sequencing-based methods have evolved to meet increasing scientific demands. We will also highlight experimental and bioinformatic challenges, aiming to provide a guide for accurate triplet repeat characterization for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Simone Maestri
- Department of Biosciences, University of Milan, Street Giovanni Celoria, 26, 20133, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Street Francesco Sforza, 35, 20122, Milan, Italy
| | - Davide Scalzo
- Department of Biosciences, University of Milan, Street Giovanni Celoria, 26, 20133, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Street Francesco Sforza, 35, 20122, Milan, Italy
| | - Gianluca Damaggio
- Department of Biosciences, University of Milan, Street Giovanni Celoria, 26, 20133, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Street Francesco Sforza, 35, 20122, Milan, Italy
| | - Martina Zobel
- Department of Biosciences, University of Milan, Street Giovanni Celoria, 26, 20133, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Street Francesco Sforza, 35, 20122, Milan, Italy
| | - Dario Besusso
- Department of Biosciences, University of Milan, Street Giovanni Celoria, 26, 20133, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Street Francesco Sforza, 35, 20122, Milan, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, Street Giovanni Celoria, 26, 20133, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare ‘Romeo ed Enrica Invernizzi’, Street Francesco Sforza, 35, 20122, Milan, Italy
| |
Collapse
|
2
|
Protic D, Polli R, Bettella E, Usdin K, Murgia A, Tassone F. Somatic Instability Leading to Mosaicism in Fragile X Syndrome and Associated Disorders: Complex Mechanisms, Diagnostics, and Clinical Relevance. Int J Mol Sci 2024; 25:13681. [PMID: 39769443 PMCID: PMC11728179 DOI: 10.3390/ijms252413681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Fragile X syndrome (FXS) is a genetic condition caused by the inheritance of alleles with >200 CGG repeats in the 5' UTR of the fragile X messenger ribonucleoprotein 1 (FMR1) gene. These full mutation (FM) alleles are associated with DNA methylation and gene silencing, which result in intellectual disabilities, developmental delays, and social and behavioral issues. Mosaicism for both the size of the CGG repeat tract and the extent of its methylation is commonly observed in individuals with the FM. Mosaicism has also been reported in carriers of premutation (PM) alleles, which have 55-200 CGG repeats. PM alleles confer risk for the fragile X premutation-associated conditions (FXPAC), including FXTAS, FXPOI, and FXAND, conditions thought to be due to the toxic consequences of transcripts containing large CGG-tracts. Unmethylated FM (UFM) alleles are transcriptionally and translationally active. Thus, they produce transcripts with toxic effects. These transcripts do produce some FMRP, the encoded product of the FMR1 gene, albeit with reduced translational efficiency. As a result, mosaicism can result in a complex clinical presentation. Here, we review the concept of mosaicism in both FXS and in PM carriers, including its potential clinical significance.
Collapse
Affiliation(s)
- Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, 11000 Belgrade, Serbia
| | - Roberta Polli
- Department of Women’s and Children’s Health, University of Padova, 35127 Padova, Italy; (R.P.); (E.B.)
- Pediatric Research Institute Città della Speranza, 35127 Padova, Italy
| | - Elisa Bettella
- Department of Women’s and Children’s Health, University of Padova, 35127 Padova, Italy; (R.P.); (E.B.)
- Pediatric Research Institute Città della Speranza, 35127 Padova, Italy
| | - Karen Usdin
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Alessandra Murgia
- Department of Women’s and Children’s Health, University of Padova, 35127 Padova, Italy; (R.P.); (E.B.)
- Pediatric Research Institute Città della Speranza, 35127 Padova, Italy
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
3
|
Loesch DZ, Chafota F, Bui MQ, Storey E, Atkinson A, Martin NG, Gordon SD, Rentería ME, Hagerman RJ, Tassone F. Parkinson's Disease Polygenic Risk Score and Neurological Involvement in Carriers of the FMR1 Premutation Allele: A Case for Genetic Modifier. Mol Genet Genomic Med 2024; 12:e70043. [PMID: 39588919 PMCID: PMC11590032 DOI: 10.1002/mgg3.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/09/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Premutation alleles of the FMR1 X-linked gene containing CGG repeat expansions ranging from 55 to 200 are associated with diverse late-onset neurological involvements, including most severe disorder termed Fragile X-associated Tremor/Ataxia Syndrome (FXTAS). It is intriguing that at least one-third of male, and a much lower than predicted from the X-linkage proportion of female carriers are free of this syndrome. This suggests the existence of secondary genetic factors modifying the risk of neurological involvements in these carriers. Considering the occasional presence of parkinsonian features in FXTAS, we explored the possibility that the Parkinson's Disease Polygenic Risk Score (PD PRS) is related to the occurrence of FXTAS or less severe neurological involvements, in premutation carriers. METHODS The Genome-wide SNP genotyping and clinical data on neurological status were obtained from 250 unrelated affected and non-affected male and female adult carriers of the premutation. The medians for the Parkinson's Disease Polygenic Risk Score (PD PRS) were compared between the groups of asymptomatic and neurologically affected carriers, and the association of PD PRS with neurological involvement in context with the other known risk factors was explored by fitting univariate and multiple logistic regression models. RESULTS There was a significant difference between the medians from the asymptomatic versus neurologically affected (FXTAS+) groups (p = 0.009). The FXTAS+ status was significantly associated with age at testing (p < 0.001), gender (p = 0.026), and with PD PRS (p = 0.021). The contribution of PD PRS remained significant after adjusting for age and gender (p = 0.044). CONCLUSIONS We have obtained the first evidence for the relationship between PD PRS and the risk of FXTAS or lesser neurological involvements in the FMR1 premutation carriers. This suggests the role of Parkinson's disease polygenic variants as genetic modifiers of the risk of late onset neurological changes in these carriers.
Collapse
Affiliation(s)
- Danuta Z. Loesch
- School of Psychology and Public HealthLa Trobe UniversityBundooraVictoriaAustralia
| | - Freddy Chafota
- Mental Health & Neuroscience ProgramQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Minh Q. Bui
- Centre for Epidemiology and Biostatistics, School of Global and Population HealthUniversity of MelbourneVictoriaAustralia
| | - Elsdon Storey
- Department of Medicine (Neuroscience)Monash University, Alfred Hospital CampusMelbourneAustralia
| | - Anna Atkinson
- School of Psychology and Public HealthLa Trobe UniversityBundooraVictoriaAustralia
| | - Nicholas G. Martin
- Mental Health & Neuroscience ProgramQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Scott D. Gordon
- Mental Health & Neuroscience ProgramQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Miguel E. Rentería
- Mental Health & Neuroscience ProgramQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Randi J. Hagerman
- Department of PediatricsUniversity of California Davis HealthSacramentoCaliforniaUSA
- Medical Investigation of Neurodevelopmental Disorders (MIND) InstituteUniversity of California Davis HealthSacramentoCaliforniaUSA
| | - Flora Tassone
- Department of Biochemistry and Molecular MedicineUniversity of CaliforniaDavisCaliforniaUSA
- School of Medicine and MIND InstituteUniversity of California Davis Medical CenterDavisCaliforniaUSA
| |
Collapse
|
4
|
Protic D, Breeze E, Mendoza G, Zafarullah M, Abbeduto L, Hagerman R, Coffey C, Cudkowicz M, Durbin-Johnson B, Ashwood P, Berry-Kravis E, Erickson CA, Filipink R, Gropman A, Lehwald L, Maxwell-Horn A, Morris S, Bennett AP, Prock L, Talboy A, Tartaglia N, Veenstra-VanderWeele J, Tassone F. Negative effect of treatment with mGluR5 negative allosteric modulator AFQ056 on blood biomarkers in young individuals with Fragile X syndrome. SAGE Open Med 2024; 12:20503121241282401. [PMID: 39483619 PMCID: PMC11526204 DOI: 10.1177/20503121241282401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/22/2024] [Indexed: 11/03/2024] Open
Abstract
Background Fragile X syndrome, with an approximate incidence rate of 1 in 4000 males to 1 in 8000 females, is the most prevalent genetic cause of heritable intellectual disability and the most common monogenic cause of autism spectrum disorder. The full mutation of the Fragile X Messenger Ribonucleoprotein-1 gene, characterized by an expansion of CGG trinucleotide repeats (>200 CGG repeats), leads to fragile X syndrome. Currently, there are no targeted treatments available for fragile X syndrome. In a recent large multi-site trial, FXLEARN, the effects of the mGluR5 negative allosteric modulator, AFQ056 (mavoglurant), were investigated, but did not show a significant impact of AFQ056 on language development in children with fragile X syndrome aged 3-6 years. Objectives The current analyses from biospecimens collected in the FXLEARN study aimed to determine whether AFQ056 affects the level of potential biomarkers associated with Akt/mTOR and matrix metalloproteinase 9 signaling in young individuals with fragile X syndrome. Previous research has indicated that these biomarkers play crucial roles in the pathophysiology of fragile X syndrome. Design A double-blind placebo-controlled parallel-group flexible-dose forced titration design. Methods Blood samples for biomarkers were collected during the FXLEARN at baseline and subsequent visits (1- and 8-month visits). Biomarker analyses included fragile X messenger ribonucleoprotein-1 genotyping by Southern blot and PCR approaches, fragile X messenger ribonucleoprotein-1 mRNA levels determined by PCR, matrix metalloproteinase 9 levels' detection using a magnetic bead panel, and targets of the Akt/mTOR signaling pathway with their phosphorylation levels detected. Results This research revealed that administering AFQ056 does not affect the expression levels of the investigated blood biomarkers in young children with fragile X syndrome. Conclusion Our findings of the lack of association between clinical improvement and biomarkers' levels in the treatment group are in line with the lack of benefit observed in the FXLEARN study. These findings indicate that AFQ056 does not provide benefits as assessed by primary or secondary endpoints. Registration ClincalTrials.gov NCT02920892.
Collapse
Affiliation(s)
- Dragana Protic
- Faculty of Medicine, Department of Pharmacology, Clinical Pharmacology, and Toxicology, University of Belgrade, Belgrade, Serbia
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia
| | - Elizabeth Breeze
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Sacramento, CA, USA
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Leonard Abbeduto
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Sacramento, CA, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | | | - Merit Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Neurological Sciences, Anatomy, and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | | | - Stephanie Morris
- Washington University Medical Center, Saint Louis Children’s Hospital, St. Louis, MO, USA
| | | | - Lisa Prock
- Boston Children’s Hospital, Boston, MA, USA
| | - Amy Talboy
- Emory University Medical Center, Atlanta, GA, USA
| | | | - Jeremy Veenstra-VanderWeele
- Center for Autism and the Developing Brain, New York-Presbyterian, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Flora Tassone
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
5
|
Maltman N, Sterling A, Santos E, Hagerman R. Language use predicts symptoms of fragile X-associated tremor/ataxia syndrome in men and women with the FMR1 premutation. Sci Rep 2024; 14:20707. [PMID: 39237554 PMCID: PMC11377817 DOI: 10.1038/s41598-024-70810-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is an age-related neurodegenerative disorder caused by a premutation of the FMR1 gene on the X chromosome. Despite the pervasive physical and cognitive effects of FXTAS, no studies have examined language in symptomatic males and females, limiting utility as an outcome measure in clinical trials of FXTAS. The goal of this work is to determine (a) the extent to which male and female FMR1 premutation carriers with FXTAS symptoms differ in their language use and (b) whether language production predicts FXTAS symptoms. Thirty-one individuals with the FMR1 premutation (21M, 10F), ages 58-85 years with some symptoms of FXTAS, were recruited from a larger cross-sectional study. Participants completed a five-minute monologic language sample. Language transcripts were assessed for rate of dysfluencies, lexical-semantics, syntax, and speech rate. Multivariable linear and ordinal regressions were used to predict FXTAS-associated symptoms, cognitive functioning, and executive functioning. Males and females did not differ in their language use. Language production predicted FXTAS symptom severity, cognitive functioning, and executive functioning. Language production difficulties may co-occur with FXTAS-associated symptoms and may be a viable outcome measure in future clinical trials, with future research needed.
Collapse
Affiliation(s)
- Nell Maltman
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI, 53705, USA.
- Department of Speech, Language, and Hearing Sciences, University of Arizona, 1131 2nd St , Tucson, AZ, 85721, USA.
| | - Audra Sterling
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI, 53705, USA
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, 1975 Willow Dr, Madison, WI, 53706, USA
| | - Ellery Santos
- MIND Institute, University of California-Davis, 2825 50th St., Sacramento, CA, 95817, USA
| | - Randi Hagerman
- MIND Institute, University of California-Davis, 2825 50th St., Sacramento, CA, 95817, USA
| |
Collapse
|
6
|
Chi MH, Bourgeois JA, Santos E, Kim K, Ponzini MD, Mendoza G, Schneider A, Hessl D, Tassone F, Hagerman RJ. Psychiatric Manifestations in Early to Middle Stages of Fragile X-Associated Tremor-Ataxia Syndrome (FXTAS). J Neuropsychiatry Clin Neurosci 2024; 37:20-28. [PMID: 39113493 DOI: 10.1176/appi.neuropsych.20230215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
OBJECTIVE The purpose of the present study was to assess the psychiatric manifestations of early to middle stages of fragile X-associated tremor-ataxia syndrome (FXTAS) and their relationship with executive function and FMR1 cytosine-guanine-guanine (CGG) repeat numbers across genders. METHODS Cross-sectional data from 100 participants (62 men, 38 women; mean±SD age=67.11±7.90 years) with FXTAS stage 1, 2, or 3 were analyzed, including demographic information, cognitive measures, psychiatric assessments (Symptom Checklist-90-Revised and Behavioral Dyscontrol Scale-II [BDS-II]), and CGG repeat number. RESULTS Participants with FXTAS stage 3 exhibited significantly worse psychiatric outcomes compared with participants with either stage 1 or 2, with distinct gender-related differences. Men showed differences in anxiety and hostility between stage 3 and combined stages 1 and 2, whereas women exhibited differences in anxiety, depression, interpersonal sensitivity, obsessive-compulsive symptoms, and somatization, as well as in the Global Severity Index, the Positive Symptom Distress Index, and the Positive Symptom Total. Among male participants, negative correlations were observed between BDS-II total scores and obsessive-compulsive symptoms, as well as between anxiety and CGG repeat number. CONCLUSIONS These findings suggest that even at early FXTAS stages, patients have significant cognitive and other psychiatric symptoms, with notable gender-specific differences. This study underscores the clinical and prognostic relevance of comorbid psychiatric conditions in FXTAS, highlighting the need for early intervention and targeted support for individuals with relatively mild motor deficits.
Collapse
Affiliation(s)
- Mei Hung Chi
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - James A Bourgeois
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Ellery Santos
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Kyoungmi Kim
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Matt Dominic Ponzini
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Guadalupe Mendoza
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Andrea Schneider
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - David Hessl
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Health, Sacramento (Chi, Santos, Kim, Ponzini, Schneider, Hessl, Tassone, Hagerman); Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan (Chi); Departments of Psychiatry and Behavioral Sciences (Bourgeois, Hessl), Pediatrics (Santos, Schneider, Hagerman), Public Health Sciences (Kim, Ponzini), and Biochemistry and Molecular Medicine (Mendoza, Tassone), University of California, Davis School of Medicine, Sacramento
| |
Collapse
|
7
|
Winarni TI, Hwang YH, Rivera SM, Hessl D, Durbin-Johnson BP, Utari A, Hagerman R, Tassone F. Apolipoproteine and KLOTHO Gene Variants Do Not Affect the Penetrance of Fragile X-Associated Tremor/Ataxia Syndrome. Int J Mol Sci 2024; 25:8103. [PMID: 39125677 PMCID: PMC11312271 DOI: 10.3390/ijms25158103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
In this study, the potential role and interaction of the APOε and KLOTHO genes on the penetrance of fragile X-associated tremor/ataxia syndrome (FXTAS) and on the IQ trajectory were investigated. FXTAS was diagnosed based on molecular, clinical and radiological criteria. Males with the premutation (PM) over 50 years, 165 with and 34 without an FXTAS diagnosis, were included in this study and were compared based on their APO (ε2-ε3-ε4) and KLOTHO variant (KL-VS) genotypes. The effect of APOε4 on FXTAS stage and on diagnosis did not differ significantly by KL-VS genotype with interaction effect p = 0.662 and p = 0.91, respectively. In the FXTAS individuals with an APOε2 allele, a marginal significance was observed towards a larger decline in verbal IQ (VIQ) in individuals with an APOε4 allele compared to those without an APOε4 allele (p = 0.071). In conclusion, our findings suggest that the APOε4 and KL-VS genotypes alone or through their interaction effect do not appear to predispose to either FXTAS diagnosis or stage in male carriers of the PM allele. A further study is needed to establish the trend of IQ decline in the FXTAS individuals who carry APOε4 with APOε2 compared to those without APOε4.
Collapse
Affiliation(s)
- Tri Indah Winarni
- Center for Biomedical Research (CEBIOR), Faculty of Medicine, Universitas Diponegoro, Semarang 50275, Central Java, Indonesia; (T.I.W.); (A.U.)
| | - Ye Hyun Hwang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - Susan M. Rivera
- Department of Psychology, University of Marlyand, College Park, MD 20742, USA;
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (D.H.); (R.H.)
| | - David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (D.H.); (R.H.)
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Blythe P. Durbin-Johnson
- Division of Biostatistics, School of Medicine, University of California Davis, Davis, CA 95616, USA;
| | - Agustini Utari
- Center for Biomedical Research (CEBIOR), Faculty of Medicine, Universitas Diponegoro, Semarang 50275, Central Java, Indonesia; (T.I.W.); (A.U.)
- Department of Pediatrics, Faculty of Medicine, Universitas Diponegoro, Semarang 50275, Central Java, Indonesia
| | - Randi Hagerman
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (D.H.); (R.H.)
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA;
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (D.H.); (R.H.)
| |
Collapse
|
8
|
Elias-Mas A, Wang JY, Rodríguez-Revenga L, Kim K, Tassone F, Hessl D, Rivera SM, Hagerman R. Enlarged perivascular spaces and their association with motor, cognition, MRI markers and cerebrovascular risk factors in male fragile X premutation carriers. J Neurol Sci 2024; 461:123056. [PMID: 38772058 DOI: 10.1016/j.jns.2024.123056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/23/2024]
Abstract
FMR1 premutation carriers (55-200 CGG repeats) are at risk of developing fragile X-associated tremor/ataxia syndrome (FXTAS), a neurodegenerative disorder associated with motor and cognitive impairment. Bilateral hyperintensities of the middle cerebellar peduncles (MCP sign) are the major radiological hallmarks of FXTAS. In the general population, enlarged perivascular spaces (PVS) are biomarkers of small vessel disease and glymphatic dysfunction and are associated with cognitive decline. Our aim was to determine if premutation carriers show higher ratings of PVS than controls and whether enlarged PVS are associated with motor and cognitive impairment, MRI features of neurodegeneration, cerebrovascular risk factors and CGG repeat length. We evaluated 655 MRIs (1-10 visits/participant) from 229 carriers (164 with FXTAS and 65 without FXTAS) and 133 controls. PVS in the basal ganglia (BG-EPVS), centrum semiovale, and midbrain were evaluated with a semiquantitative scale. Mixed-effects models were used for statistical analysis adjusting for age. In carriers with FXTAS, we revealed that (1) BG-PVS ratings were higher than those of controls and carriers without FXTAS; (2) BG-PVS severity was associated with brain atrophy, white matter hyperintensities, enlarged ventricles, FXTAS stage and abnormal gait; (3) age-related increase in BG-PVS was associated with cognitive dysfunction; and (4) PVS ratings of all three regions showed robust associations with CGG repeat length and were higher in carriers with the MCP sign than carriers without the sign. This study demonstrates clinical relevance of PVS in FXTAS especially in the basal ganglia region and suggests microangiopathy and dysfunctional cerebrospinal fluid circulation in FXTAS physiopathology.
Collapse
Affiliation(s)
- Andrea Elias-Mas
- Radiology Department, Hospital Universitari Mútua de Terrassa, Terrassa, Barcelona, Spain; Institute for Research and Innovation Parc Taulí (I3PT), Sabadell, Spain; Genetics Doctorate Program, Universitat de Barcelona (UB), Barcelona, Spain.
| | - Jun Yi Wang
- Center for Mind and Brain, University of California Davis, CA, United States.
| | - Laia Rodríguez-Revenga
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, Barcelona, Spain; CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain; Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Kyoungmi Kim
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, United States.
| | - Flora Tassone
- MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States.
| | - David Hessl
- MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States.
| | - Susan M Rivera
- Center for Mind and Brain, University of California Davis, CA, United States; MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Psychology, University of Maryland, College Park, MD, United States.
| | - Randi Hagerman
- MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Pediatrics, University of California Davis Medical Center, Sacramento, CA, United States.
| |
Collapse
|
9
|
Seng P, Montanaro FAM, Biag HMB, Salcedo-Arellano MJ, Kim K, Ponzini MD, Tassone F, Schneider A, Abbeduto L, Thurman AJ, Hessl D, Bolduc FV, Jacquemont S, Lippé S, Hagerman RJ. Longitudinal follow-up of metformin treatment in Fragile X Syndrome. Front Psychol 2024; 15:1305597. [PMID: 38939222 PMCID: PMC11210589 DOI: 10.3389/fpsyg.2024.1305597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 05/22/2024] [Indexed: 06/29/2024] Open
Abstract
Introduction Metformin has been used as a targeted treatment to potentially improve cognition and slow the typical IQ decline that occurs during development among individuals with fragile X syndrome (FXS). In this follow-up study, we are following the trajectory of IQ and adaptive behavior changes over 1 to 3 years in individuals with FXS who are clinically treated with metformin in an open label trial. Method Individuals with FXS ages 6 to 25 years (mean 13.15 ± 5.50) and nonverbal IQ mean 57.69 (±15.46) were treated for 1-3 years (1.88 ± 0.63). They all had a baseline IQ test using the Leiter-III non-verbal cognitive assessment and the Vineland-III adaptive behavior assessment before the start of metformin. Repeat Leiter-III and Vineland-III were completed after at least 1 year of metformin (500-1,000 mg/dose given twice a day). Result There were no significant changes in non-verbal IQ or in the adaptive behavior measurements at FDR < 0.05. The findings thus far indicate that both IQ and adaptive behavior are stable over time, and we did not see a significant decline in either measure. Conclusion Overall, the small sample size and short follow-up duration limit the interpretation of the effects of metformin on cognitive development and adaptive functioning. There is individual variability but overall for the group there was no significant decline in IQ or adaptive behavior.
Collapse
Affiliation(s)
- Panhaneath Seng
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
| | - Federica Alice Maria Montanaro
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Education, Psychology, Communication, University of Bari Aldo Moro, Bari, Italy
| | - Hazel Maridith Barlahan Biag
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Maria Jimena Salcedo-Arellano
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Kyoungmi Kim
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
- Integrative Genetics and Genomics Graduate Group, University of California Davis, Davis, CA, United States
| | - Matthew Dominic Ponzini
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Flora Tassone
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Andrea Schneider
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Leonard Abbeduto
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Angela John Thurman
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - David Hessl
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Francois V. Bolduc
- Department of Pediatrics, Department of Medical Genetics, Women and Children Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Sebastien Jacquemont
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Sarah Lippé
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Randi J. Hagerman
- MIND Institute, University of California Davis Health System, Sacramento, CA, United States
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
10
|
Loesch DZ, Atkinson A, Hall DA, Tassone F, Stimpson P, Storey E. Cognitive status correlates of subclinical action tremor in female carriers of FMR1 premutation. Front Neurol 2024; 15:1401286. [PMID: 38903175 PMCID: PMC11188871 DOI: 10.3389/fneur.2024.1401286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Background There is evidence for a significant excess of kinetic upper limb tremor in non-FXTAS female FMR1 premutation carriers. The present study explores the possibility that this tremor is associated with various other features reminiscent of those occurring in syndromic FXTAS. Sample/methods This study analyzed the data from an Australian cohort of 48 asymptomatic premutation women. We utilized spiral drawings from CRST, representing action tremor; the CRST total tremor; and ICARS- kinetic tremors/cerebellar ataxia scales. Cognitive tests (involving executive functioning) included SDMT, TMT, two subtests of the WAIS-III: MR and Similarities. Spearman Rank correlations assessed the relationships between the above measures, and the Chi-square tested hypothesis about the association between the white matter hyperintensities (wmhs) in the splenium of corpus callosum assessed from MR images and spiral drawings scores. Results The spiral drawing scores were significantly correlated with all three non-verbal cognitive test scores, and with the CRST scores; the latter correlated with all four cognitive test measures. Similarities (verbal) scores correlated with CRST, ICARS, and with the remaining cognitive scores. Ordered spiral scores' categories were significantly associated with the degree of splenium involvement. Conclusion This study showed that, in non-FXTAS premutation female carriers, sub-symptomatic forms of kinetic tremor were associated with a broader motor, and cognitive (especially executive) dysfunction.
Collapse
Affiliation(s)
- Danuta Z. Loesch
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - Anna Atkinson
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - Deborah A. Hall
- Department of Neurological Sciences, Rush University Medical Centre, Chicago, IL, United States
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
- MIND Institute, University of California Davis Medical Centre, Davis, CA, United States
| | - Paige Stimpson
- Wellness and Recovery Centre, Monash Medical Centre, Clayton, VIC, Australia
| | - Elsdon Storey
- Department of Medicine (Neuroscience), Alfred Hospital Campus, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Jiraanont P, Zafarullah M, Sulaiman N, Espinal GM, Randol JL, Durbin-Johnson B, Schneider A, Hagerman RJ, Hagerman PJ, Tassone F. FMR1 Protein Expression Correlates with Intelligence Quotient in Both Peripheral Blood Mononuclear Cells and Fibroblasts from Individuals with an FMR1 Mutation. J Mol Diagn 2024; 26:498-509. [PMID: 38522837 DOI: 10.1016/j.jmoldx.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Fragile X syndrome (FXS) is the most common heritable form of intellectual disability and is caused by CGG repeat expansions exceeding 200 (full mutation). Such expansions lead to hypermethylation and transcriptional silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene. As a consequence, little or no FMR1 protein (FMRP) is produced; absence of the protein, which normally is responsible for neuronal development and maintenance, causes the syndrome. Previous studies have demonstrated the causal relationship between FMRP levels and cognitive abilities in peripheral blood mononuclear cells (PBMCs) and dermal fibroblast cell lines of patients with FXS. However, it is arguable whether PBMCs or fibroblasts would be the preferred surrogate for measuring molecular markers, particularly FMRP, to represent the cognitive impairment, a core symptom of FXS. To address this concern, CGG repeats, methylation status, FMR1 mRNA, and FMRP levels were measured in both PBMCs and fibroblasts derived from 66 individuals. The findings indicated a strong association between FMR1 mRNA expression levels and CGG repeat numbers in PBMCs of premutation males after correcting for methylation status. Moreover, FMRP expression levels from both PBMCs and fibroblasts of male participants with a hypermethylated full mutation and with mosaicism demonstrated significant association between the intelligence quotient levels and FMRP levels, suggesting that PBMCs may be preferable for FXS clinical studies, because of their greater accessibility.
Collapse
Affiliation(s)
- Poonnada Jiraanont
- Division of Molecular and Cellular Medicine, Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California
| | - Noor Sulaiman
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California
| | - Glenda M Espinal
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California
| | - Jamie L Randol
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California
| | - Blythe Durbin-Johnson
- Division of Biostatistics, University of California, Davis, School of Medicine, Davis, California
| | - Andrea Schneider
- Department of Pediatrics, University of California, Davis, School of Medicine, Davis, California; UC Davis MIND Institute, University of California, Davis, Sacramento, California
| | - Randi J Hagerman
- Department of Pediatrics, University of California, Davis, School of Medicine, Davis, California; UC Davis MIND Institute, University of California, Davis, Sacramento, California
| | - Paul J Hagerman
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California; UC Davis MIND Institute, University of California, Davis, Sacramento, California
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California; UC Davis MIND Institute, University of California, Davis, Sacramento, California.
| |
Collapse
|
12
|
Randol JL, Kim K, Ponzini MD, Tassone F, Falcon AK, Hagerman RJ, Hagerman PJ. Variation of FMRP Expression in Peripheral Blood Mononuclear Cells from Individuals with Fragile X Syndrome. Genes (Basel) 2024; 15:356. [PMID: 38540415 PMCID: PMC10969917 DOI: 10.3390/genes15030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 04/07/2024] Open
Abstract
Fragile X syndrome (FXS) is the most common heritable cause of intellectual disability and autism spectrum disorder. The syndrome is often caused by greatly reduced or absent protein expression from the fragile X messenger ribonucleoprotein 1 (FMR1) gene due to expansion of a 5'-non-coding trinucleotide (CGG) element beyond 200 repeats (full mutation). To better understand the complex relationships among FMR1 allelotype, methylation status, mRNA expression, and FMR1 protein (FMRP) levels, FMRP was quantified in peripheral blood mononuclear cells for a large cohort of FXS (n = 154) and control (n = 139) individuals using time-resolved fluorescence resonance energy transfer. Considerable size and methylation mosaicism were observed among individuals with FXS, with FMRP detected only in the presence of such mosaicism. No sample with a minimum allele size greater than 273 CGG repeats had significant levels of FMRP. Additionally, an association was observed between FMR1 mRNA and FMRP levels in FXS samples, predominantly driven by those with the lowest FMRP values. This study underscores the complexity of FMR1 allelotypes and FMRP expression and prompts a reevaluation of FXS therapies aimed at reactivating large full mutation alleles that are likely not capable of producing sufficient FMRP to improve cognitive function.
Collapse
Affiliation(s)
- Jamie L. Randol
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Kyoungmi Kim
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, UC Davis Health, Sacramento, CA 95817, USA
- Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Matthew D. Ponzini
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, UC Davis Health, Sacramento, CA 95817, USA
- Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, UC Davis Health, Sacramento, CA 95817, USA
| | - Alexandria K. Falcon
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, UC Davis Health, Sacramento, CA 95817, USA
- Department of Pediatrics, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Paul J. Hagerman
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, UC Davis Health, Sacramento, CA 95817, USA
| |
Collapse
|
13
|
Tak Y, Schneider A, Santos E, Randol JL, Tassone F, Hagerman P, Hagerman RJ. Unmethylated Mosaic Full Mutation Males without Fragile X Syndrome. Genes (Basel) 2024; 15:331. [PMID: 38540390 PMCID: PMC10970065 DOI: 10.3390/genes15030331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 04/07/2024] Open
Abstract
Fragile X syndrome (FXS) is the leading inherited cause of intellectual disability (ID) and single gene cause of autism. Although most patients with FXS and the full mutation (FM) have complete methylation of the fragile X messenger ribonucleoprotein 1 (FMR1) gene, some have mosaicism in methylation and/or CGG repeat size, and few have completely unmethylated FM alleles. Those with a complete lack of methylation are rare, with little literature about the cognitive and behavioral phenotypes of these individuals. A review of past literature was conducted regarding individuals with unmethylated and mosaic FMR1 FM. We report three patients with an unmethylated FM FMR1 alleles without any behavioral or cognitive deficits. This is an unusual presentation for men with FM as most patients with an unmethylated FM and no behavioral phenotypes do not receive fragile X DNA testing or a diagnosis of FXS. Our cases showed that mosaic males with unmethylated FMR1 FM alleles may lack behavioral phenotypes due to the presence of smaller alleles producing the FMR1 protein (FMRP). However, these individuals could be at a higher risk of developing fragile X-associated tremor/ataxia syndrome (FXTAS) due to the increased expression of mRNA, similar to those who only have a premutation.
Collapse
Affiliation(s)
- YeEun Tak
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (Y.T.); (E.S.); (F.T.); (P.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Andrea Schneider
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Ellery Santos
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (Y.T.); (E.S.); (F.T.); (P.H.)
| | - Jamie Leah Randol
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (Y.T.); (E.S.); (F.T.); (P.H.)
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Paul Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (Y.T.); (E.S.); (F.T.); (P.H.)
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (Y.T.); (E.S.); (F.T.); (P.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| |
Collapse
|
14
|
Juarez P, Salcedo-Arellano MJ, Dufour B, Martinez-Cerdeño V. Fragile X cortex is characterized by decreased parvalbumin-expressing interneurons. Cereb Cortex 2024; 34:bhae103. [PMID: 38521994 PMCID: PMC10960956 DOI: 10.1093/cercor/bhae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/25/2024] Open
Abstract
Fragile X syndrome is a genetic neurodevelopmental disorder caused by a mutation of the fragile X messenger ribonucleoprotein 1 (FMR1) gene in the X chromosome. Many fragile X syndrome cases present with autism spectrum disorder and fragile X syndrome cases account for up to 5% of all autism spectrum disorder cases. The cellular composition of the fragile X syndrome cortex is not well known. We evaluated alterations in the number of Calbindin, Calretinin, and Parvalbumin expressing interneurons across 5 different cortical areas, medial prefrontal cortex (BA46), primary somatosensory cortex (BA3), primary motor cortex (BA4), superior temporal cortex (BA22), and anterior cingulate cortex (BA24) of fragile X syndrome and neurotypical brains. Compared with neurotypical cases, fragile X syndrome brains displayed a significant reduction in the number of PV+ interneurons in all areas and of CR+ interneurons in BA22 and BA3. The number of CB+ interneurons did not differ. These findings are the first to demonstrate that fragile X syndrome brains are characterized by cortical wide PV+ interneuron deficits across multiple cortical areas. These add to the idea that deficits in PV+ interneurons could disrupt the cortical balance and promote clinical deficits in fragile X syndrome patients and help to develop novel therapies for neurodevelopmental disorders like fragile X syndrome and autism spectrum disorder.
Collapse
Affiliation(s)
- Pablo Juarez
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
| | - Maria Jimena Salcedo-Arellano
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
| | - Brett Dufour
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
| | - Veronica Martinez-Cerdeño
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
| |
Collapse
|
15
|
Santos E, Clark C, Biag HMB, Tang SJ, Kim K, Ponzini MD, Schneider A, Giulivi C, Montanaro FAM, Gipe JTE, Dayton J, Randol JL, Yao PJ, Manolopoulos A, Kapogiannis D, Hwang YH, Hagerman P, Hagerman R, Tassone F. Open-Label Sulforaphane Trial in FMR1 Premutation Carriers with Fragile-X-Associated Tremor and Ataxia Syndrome (FXTAS). Cells 2023; 12:2773. [PMID: 38132093 PMCID: PMC10741398 DOI: 10.3390/cells12242773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/03/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Fragile X (FMR1) premutation is a common mutation that affects about 1 in 200 females and 1 in 450 males and can lead to the development of fragile-X-associated tremor/ataxia syndrome (FXTAS). Although there is no targeted, proven treatment for FXTAS, research suggests that sulforaphane, an antioxidant present in cruciferous vegetables, can enhance mitochondrial function and maintain redox balance in the dermal fibroblasts of individuals with FXTAS, potentially leading to improved cognitive function. In a 24-week open-label trial involving 15 adults aged 60-88 with FXTAS, 11 participants successfully completed the study, demonstrating the safety and tolerability of sulforaphane. Clinical outcomes and biomarkers were measured to elucidate the effects of sulforaphane. While there were nominal improvements in multiple clinical measures, they were not significantly different after correction for multiple comparisons. PBMC energetic measures showed that the level of citrate synthase was higher after sulforaphane treatment, resulting in lower ATP production. The ratio of complex I to complex II showed positive correlations with the MoCA and BDS scores. Several mitochondrial biomarkers showed increased activity and quantity and were correlated with clinical improvements.
Collapse
Affiliation(s)
- Ellery Santos
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95817, USA
| | - Courtney Clark
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95817, USA
| | - Hazel Maridith B. Biag
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95817, USA
| | - Si Jie Tang
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95817, USA
| | - Kyoungmi Kim
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Matthew D. Ponzini
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Andrea Schneider
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95817, USA
| | - Cecilia Giulivi
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Federica Alice Maria Montanaro
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
- Department of Education, Psychology, Communication, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Jesse Tran-Emilia Gipe
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Jacquelyn Dayton
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Jamie L. Randol
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95616, USA
| | - Pamela J. Yao
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 212241, USA (A.M.); (D.K.)
| | - Apostolos Manolopoulos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 212241, USA (A.M.); (D.K.)
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 212241, USA (A.M.); (D.K.)
| | - Ye Hyun Hwang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95616, USA
| | - Paul Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95616, USA
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95817, USA
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis Health, Sacramento, CA 95817, USA (A.S.); (R.H.)
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
16
|
Aishworiya R, Tak Y, Ponzini MD, Biag HMB, Salcedo-Arellano MJ, Kim K, Tassone F, Schneider A, Thurman AJ, Abbeduto L, Hessl D, Randol JL, Bolduc FV, Lippe S, Hagerman P, Hagerman R. Adaptive, behavioral, and cognitive outcomes in individuals with fragile X syndrome with varying autism severity. Int J Dev Neurosci 2023; 83:715-727. [PMID: 37724826 PMCID: PMC10868665 DOI: 10.1002/jdn.10299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/06/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
This study aimed to determine the association between severity of autism spectrum disorder (ASD) and cognitive, behavioral, and molecular measures in individuals with fragile X syndrome (FXS). Study inclusion criteria included individuals with FXS and (1) age 6-40 years, (2) full-scale IQ < 84, and (3) language ≥3-word phrases. ASD symptom severity was determined by Autism Diagnostic Observation Schedule-2 (ADOS-2). Other measures identified non-verbal IQ, adaptive skills, and aberrant behaviors. Molecular measures included blood FMR1 and CYFIP1 mRNA levels, FMRP and MMP9 levels. Analysis of variance (ANOVA) and Spearman's correlations were used to compare ASD severity groups. Data from 54 individuals was included with no/mild (N = 7), moderate (N = 18), and severe (N = 29) ASD. Individuals with high ASD severity had lower adaptive behavior scores (47.48 ± 17.49) than the no/mild group (69.00 ± 20.45, p = 0.0366); they also had more challenging behaviors, lethargy, and stereotypic behaviors. CYFIP1 mRNA expression levels positively correlated with the ADOS-2 comparison score(r2 = 0.33, p = 0.0349), with no significant correlations with other molecular markers. In conclusion, autism symptom severity is associated with more adverse cognitive and adaptive skills and specific behaviors in FXS, whereas CYFIP1 mRNA expression levels may be a potential biomarker for severity of ASD in FXS.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - YeEun Tak
- University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Matthew Dominic Ponzini
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Hazel Maridith Barlahan Biag
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Kyoungmi Kim
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Andrea Schneider
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Angela John Thurman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Leonard Abbeduto
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - David Hessl
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Jamie Leah Randol
- University of California Davis School of Medicine, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California, United States of America
- Integrative Genetics and Genomics Graduate Group, University of California Davis, One Shields Avenue, Davis, California, United States of America
- UC Davis Biotechnology Program, University of California Davis, Davis, California, United States of America
| | - Francois V Bolduc
- Division of Pediatric Neurology, Pediatrics, University of Alberta, Alberta, Canada
- Division of Medical Genetics, University of Alberta, Alberta, Canada
| | - Sarah Lippe
- Département de Psychologie, Université de Montréal, Québec, Canada
- CHU Sainte-Justine Research Center, Université de Montréal, Québec, Canada
| | - Paul Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- University of California Davis School of Medicine, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| |
Collapse
|
17
|
Bekheet MHY, Mansour LA, Elkaffas RH, Kamel MA, Elmonem MA. Serum matrix metalloproteinase-9 (MMP9) and amyloid-beta protein precursor (APP) as potential biomarkers in children with Fragile-X syndrome: A cross sectional study. Clin Biochem 2023; 121-122:110659. [PMID: 37797798 DOI: 10.1016/j.clinbiochem.2023.110659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Fragile-X syndrome(FXS) is a neurological disease caused by abnormal repeats in the 5'untranslated region of the FMR1 gene leading to a defective fragile-X-messenger-ribonucleoprotein-1 (FMRP). Although relatively common in children, it is usually under-diagnosed especially in developing countries where genetic screening is not routinely practiced. So far, FXS lacks a laboratory biomarker that can be used for screening, severity scoring or therapeutic monitoring of potential new treatments. METHODS 110 subjects were recruited; 80 male children with suspected FXS and 30 matched healthy children. We evaluated the clinical utility of serum matrix metalloproteinase-9(MMP9) and amyloid-beta protein precursor(APP) as potential biomarkers for FXS. RESULTS Out of 80 suspected children, 14 had full mutation, 8 had the premutation and 58 children had normal genotypes. No statistically-significant difference was detected between children with different genotypes concerning age of onset(P = 0.658), main clinical presentation(P = 0.388), clinical severity-score(P = 0.799), patient's disease-course(P = 0.719) and intellectual disability(P = 0.351). Both MMP9 and APP showed a statistically significant difference when comparing different genotype subgroups(P = 0.019 and < 0.001, respectively). Clinically, MMP9 levels were highest in children presenting with language defects, while APP was highest in children with neurodevelopmental delay. In receiver operating curve analysis, comparing full and premutation with the normal genotype group, MMP9 has an area-under-the-curve of 0.701(95 % CI 0.557-0.845), while APP was marginally better at 0.763(95 % CI 0.620-0.906). When combined together, elevated MMP9 or APP had excellent sensitivity > 95 % for picking-up FXS cases in the clinical setting. CONCLUSIONS Screening for circulating biomarkers in the absence of FXS genetic diagnosis is justified. Our study is the first to evaluate both MMP9 and APP in FXS suspected children in a clinical setting and to assess their correlation with disease presentation and severity.
Collapse
Affiliation(s)
- Mohamed H Y Bekheet
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Lamiaa A Mansour
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rasha H Elkaffas
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona A Kamel
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed A Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
18
|
Hou F, Mao A, Shan S, Li Y, Meng W, Zhan J, Nie W, Jin H. Evaluating the clinical utility of a long-read sequencing-based approach in genetic testing of fragile-X syndrome. Clin Chim Acta 2023; 551:117614. [PMID: 38375623 DOI: 10.1016/j.cca.2023.117614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/19/2023] [Accepted: 10/22/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Fragile X syndrome (FXS) arises from the FMR1 CGG expansion. Comprehensive genetic testing for FMR1 CGG expansions, AGG interruptions, and microdeletions is essential to provide genetic counseling for females carrying premutation alleles. However, conventional PCR-based FMR1 assays mainly focus on CGG repeats, and could detect AGG interruption only in males. METHODS The clinical utility of a long-read sequencing-based assay termed comprehensive analysis of FXS (CAFXS) was evaluated in 238 high-risk samples by comparing to conventional PCR assays. RESULTS PCR assays identified five premuation and three full mutation categories alleles in all the samples, and CAFXS successfully called all the FMR1 CGG expansion. CAFXS identified 24-bp microdeletions upstream to the trinucleotide region with 30 CGG repeats, which was miscalled by the length-based PCR methods. CAFXS also identified a 187-bp deletion in about 1/7 of the sequencing reads in a male patient with mosaic full mutation alleles. CAFXS allowed for precise constructing the FMR1 CGG repeat and AGG interruption pattern in all the samples, and identified a novel and alternative CGA interruption in one normal female sample. CONCLUSIONS CAFXS represents a more comprehensive and accurate approach for FXS genetic testing that potentially enables more informed genetic counseling compared to PCR-based methods.
Collapse
Affiliation(s)
- Fei Hou
- Department of Prenatal Diagnosis, Jinan Maternal and Child Health Hospital, Jinan 250001, Shandong Province, China
| | - Aiping Mao
- Berry Genomics Corporation, Beijing 102200, China
| | - Shan Shan
- Department of Prenatal Diagnosis, Jinan Maternal and Child Health Hospital, Jinan 250001, Shandong Province, China
| | - Yan Li
- Department of Prenatal Diagnosis, Jinan Maternal and Child Health Hospital, Jinan 250001, Shandong Province, China
| | - Wanli Meng
- Berry Genomics Corporation, Beijing 102200, China
| | - Jiahan Zhan
- Berry Genomics Corporation, Beijing 102200, China
| | - Wenying Nie
- Department of Prenatal Diagnosis, Jinan Maternal and Child Health Hospital, Jinan 250001, Shandong Province, China
| | - Hua Jin
- Department of Prenatal Diagnosis, Jinan Maternal and Child Health Hospital, Jinan 250001, Shandong Province, China.
| |
Collapse
|
19
|
Dy ABC, Tanchanco LBS, Sy JCY, Levantino MD, Hagerman RJ. Screening for Fragile X Syndrome Among Filipino Children with Autism Spectrum Disorder. J Autism Dev Disord 2023; 53:4465-4473. [PMID: 35972625 DOI: 10.1007/s10803-022-05707-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Individuals with autism spectrum disorder present with difficulties in social communication, restricted interests or behaviors and other co-morbidities. About 2 to 10% of cases of autism have a genetic cause, and Fragile X Syndrome (FXS) is reported in 0 to 6.5% of individuals with autism. However, the FXS and premutation prevalence among Filipino children has never been reported. The aim of the study was to establish the presence of FXS or premutation carriers among Filipino children with autism and to describe the phenotypic characteristic of cases identified. Blood was collected from 235 children aged 2-6 years old and diagnosed with autism. Samples were analyzed using PCR methods to amplify CGG repeats in the FMRI gene. The diagnosis of autism was confirmed through the Autism Diagnostic Observation Schedule-2. Additional characteristics were documented from a physical examination, Griffiths Scales of Child Development assessment and a parent-answered questionnaire using the Vineland Adaptive Behavior Scale. Fragile X testing through PCR methods in 235 children with diagnosed autism showed 220 (93.6%) were negative, no full mutations, 1 (0.436%) premutation carrier and 14 (5.95%) cases contained intermediate alleles. The FXS testing was limited to confirmed cases of autism, which is considered a high-risk group and does not provide prevalence for the general Filipino population. Subjects were self-referred or referred by clinicians, which may not represent the Filipino autism population with a bias towards those with means for clinical consultations and ability to travel to the place of testing. Samples were not measured for mosaicism, DNA methylation or AGG interspersion patterns. These may have effects on the CGG repeat expansion and overall presentation of FXS. Findings from a single premutation carrier cannot characterize features distinctly present in Filipinos with the mutation. Nevertheless, these results support the data that the prevalence of FXS in Asian populations may be lower than non-Asian populations. This can contribute to a better understanding of FXS and genetic causes of autism in the Philippines and other Asian populations.
Collapse
Affiliation(s)
- Angel Belle C Dy
- Ateneo de Manila University School of Medicine and Public Health, Don Eugenio Lopez Sr. Medical Complex, Pasig City, NCR, Philippines.
| | - Lourdes Bernadette S Tanchanco
- Ateneo de Manila University School of Medicine and Public Health, Don Eugenio Lopez Sr. Medical Complex, Pasig City, NCR, Philippines
| | - Jenica Clarisse Y Sy
- Ateneo de Manila University School of Medicine and Public Health, Don Eugenio Lopez Sr. Medical Complex, Pasig City, NCR, Philippines
| | - Myla Dominicina Levantino
- Ateneo de Manila University School of Medicine and Public Health, Don Eugenio Lopez Sr. Medical Complex, Pasig City, NCR, Philippines
| | - Randi J Hagerman
- University of California Davis, MIND Institute, Sacramento, CA, USA
| |
Collapse
|
20
|
Ziaei Jam H, Li Y, DeVito R, Mousavi N, Ma N, Lujumba I, Adam Y, Maksimov M, Huang B, Dolzhenko E, Qiu Y, Kakembo FE, Joseph H, Onyido B, Adeyemi J, Bakhtiari M, Park J, Javadzadeh S, Jjingo D, Adebiyi E, Bafna V, Gymrek M. A deep population reference panel of tandem repeat variation. Nat Commun 2023; 14:6711. [PMID: 37872149 PMCID: PMC10593948 DOI: 10.1038/s41467-023-42278-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023] Open
Abstract
Tandem repeats (TRs) represent one of the largest sources of genetic variation in humans and are implicated in a range of phenotypes. Here we present a deep characterization of TR variation based on high coverage whole genome sequencing from 3550 diverse individuals from the 1000 Genomes Project and H3Africa cohorts. We develop a method, EnsembleTR, to integrate genotypes from four separate methods resulting in high-quality genotypes at more than 1.7 million TR loci. Our catalog reveals novel sequence features influencing TR heterozygosity, identifies population-specific trinucleotide expansions, and finds hundreds of novel eQTL signals. Finally, we generate a phased haplotype panel which can be used to impute most TRs from nearby single nucleotide polymorphisms (SNPs) with high accuracy. Overall, the TR genotypes and reference haplotype panel generated here will serve as valuable resources for future genome-wide and population-wide studies of TRs and their role in human phenotypes.
Collapse
Affiliation(s)
- Helyaneh Ziaei Jam
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Yang Li
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ross DeVito
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Nima Mousavi
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, USA
| | - Nichole Ma
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ibra Lujumba
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Yagoub Adam
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Mikhail Maksimov
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Bonnie Huang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | | | - Yunjiang Qiu
- Illumina Incorporated, San Diego, CA, 92122, USA
| | - Fredrick Elishama Kakembo
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Habi Joseph
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Blessing Onyido
- Department of Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Jumoke Adeyemi
- Department of Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Mehrdad Bakhtiari
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Jonghun Park
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Sara Javadzadeh
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Daudi Jjingo
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Department of Computer Science, Makerere University, Kampala, Uganda
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun, 112233, Nigeria
- Department of Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun, 112233, Nigeria
- Applied Bioinformatics Division, German Cancer Research Center (DKFZ), Heidelberg, Baden-Württemberg, 69120, Germany
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Melissa Gymrek
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
21
|
Aishworiya R, Biag HMB, Salcedo-Arellano MJ, Musa Z, Schneider A, Clark C, Santos E, Tassone F, Hagerman R. Fragile X Syndrome and Fetal Alcohol Syndrome: Occurrence of Dual Diagnosis in a Set of Triplets. J Dev Behav Pediatr 2023; 44:e470-e475. [PMID: 37556593 PMCID: PMC10527597 DOI: 10.1097/dbp.0000000000001204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 06/05/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Fragile X syndrome (FXS) and fetal alcohol syndrome disorders are both common causes of intellectual disability in children. When both conditions are present in the same individual, the resultant phenotype may make identification of clinical issues and management challenging. CASE PRESENTATION In this case report, we present a case of triplets who had significant in utero alcohol exposure; 2 of whom also have FXS and the other not having the fragile X mutation. The siblings with FXS have subtle differences in the physical phenotype compared with the other one, who has prominent features of partial fetal alcohol syndrome instead. However, all 3 siblings have intellectual impairment (although this is more severe in the 2 with FXS), meet diagnostic criteria for autism spectrum disorder, and present with severe behavioral challenges. The clinical presentation of the 2 siblings with FXS is much more severe as compared to a child with FXS alone, and this is likely due to the additive effect of in utero alcohol exposure and environmental factors. We discuss the combination of these 2 pathologies and how this can affect the overall clinical presentation. CONCLUSION In the management of children with FXS, evaluation for other risk factors that can have neurobehavioral sequelae is important, and these can affect clinical presentation and prognosis.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hazel Maridith Barlahan Biag
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Zayan Musa
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Andrea Schneider
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Courtney Clark
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Ellery Santos
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| |
Collapse
|
22
|
Zafarullah M, Li J, Salemi MR, Phinney BS, Durbin-Johnson BP, Hagerman R, Hessl D, Rivera SM, Tassone F. Blood Proteome Profiling Reveals Biomarkers and Pathway Alterations in Fragile X PM at Risk for Developing FXTAS. Int J Mol Sci 2023; 24:13477. [PMID: 37686279 PMCID: PMC10488017 DOI: 10.3390/ijms241713477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Fragile X-associated Tremor/Ataxia Syndrome (FXTAS) is a neurodegenerative disorder associated with the FMR1 premutation. Currently, it is not possible to determine when and if individual premutation carriers will develop FXTAS. Thus, with the aim to identify biomarkers for early diagnosis, development, and progression of FXTAS, along with associated dysregulated pathways, we performed blood proteomic profiling of premutation carriers (PM) who, as part of an ongoing longitudinal study, emerged into two distinct groups: those who developed symptoms of FXTAS (converters, CON) over time (at subsequent visits) and those who did not (non-converters, NCON). We compared these groups to age-matched healthy controls (HC). We assessed CGG repeat allele size by Southern blot and PCR analysis. The proteomic profile was obtained by liquid chromatography mass spectrometry (LC-MS/MS). We identified several significantly differentiated proteins between HC and the PM groups at Visit 1 (V1), Visit 2 (V2), and between the visits. We further reported the dysregulated protein pathways, including sphingolipid and amino acid metabolism. Our findings are in agreement with previous studies showing that pathways involved in mitochondrial bioenergetics, as observed in other neurodegenerative disorders, are significantly altered and appear to contribute to the development of FXTAS. Lastly, we compared the blood proteome of the PM who developed FXTAS over time with the CSF proteome of the FXTAS patients recently reported and found eight significantly differentially expressed proteins in common. To our knowledge, this is the first report of longitudinal proteomic profiling and the identification of unique biomarkers and dysregulated protein pathways in FXTAS.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - Jie Li
- Genome Center, Bioinformatics Core, University of California Davis, Davis, CA 95616, USA;
| | - Michelle R. Salemi
- Genome Center, Proteomics Core, Genome and Biomedical Sciences Facility, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (B.S.P.)
| | - Brett S. Phinney
- Genome Center, Proteomics Core, Genome and Biomedical Sciences Facility, University of California Davis, Davis, CA 95616, USA; (M.R.S.); (B.S.P.)
| | - Blythe P. Durbin-Johnson
- Division of Biostatistics, School of Medicine, University of California Davis, Davis, CA 95616, USA;
| | - Randi Hagerman
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.H.); (D.H.); (S.M.R.)
- Department of Pediatrics, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.H.); (D.H.); (S.M.R.)
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Susan M. Rivera
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.H.); (D.H.); (S.M.R.)
- Department of Psychology, University of California Davis, Davis, CA 95616, USA
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA;
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.H.); (D.H.); (S.M.R.)
| |
Collapse
|
23
|
Salcedo-Arellano MJ, Johnson MD, McLennan YA, Hwang YH, Juarez P, McBride EL, Pantoja AP, Durbin-Johnson B, Tassone F, Hagerman RJ, Martínez-Cerdeño V. Brain Metabolomics in Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). Cells 2023; 12:2132. [PMID: 37681866 PMCID: PMC10487256 DOI: 10.3390/cells12172132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023] Open
Abstract
The course of pathophysiological mechanisms involved in fragile X-associated tremor/ataxia syndrome (FXTAS) remains largely unknown. Previous proteomics and metabolomics studies conducted in blood samples collected from FMR1 premutation carriers with FXTAS reported abnormalities in energy metabolism, and precursors of gluconeogenesis showed significant changes in plasma expression levels in FMR1 premutation carriers who developed FXTAS. We conducted an analysis of postmortem human brain tissues from 44 donors, 25 brains with FXTAS, and 19 matched controls. We quantified the metabolite relative abundance in the inferior temporal gyrus and the cerebellum using untargeted mass spectrometry (MS)-based metabolomics. We investigated how the metabolite type and abundance relate to the number of cytosine-guanine-guanine (CGG) repeats, to markers of neurodegeneration, and to the symptoms of FXTAS. A metabolomic analysis identified 191 primary metabolites, the data were log-transformed and normalized prior to the analysis, and the relative abundance was compared between the groups. The changes in the relative abundance of a set of metabolites were region-specific with some overlapping results; 22 metabolites showed alterations in the inferior temporal gyrus, while 21 showed differences in the cerebellum. The relative abundance of cytidine was decreased in the inferior temporal gyrus, and a lower abundance was found in the cases with larger CGG expansions; oleamide was significantly decreased in the cerebellum. The abundance of 11 metabolites was influenced by changes in the CGG repeat number. A histological evaluation found an association between the presence of microhemorrhages in the inferior temporal gyrus and a lower abundance of 2,5-dihydroxypyrazine. Our study identified alterations in the metabolites involved in the oxidative-stress response and bioenergetics in the brains of individuals with FXTAS. Significant changes in the abundance of cytidine and oleamide suggest their potential as biomarkers and therapeutic targets for FXTAS.
Collapse
Affiliation(s)
- Maria Jimena Salcedo-Arellano
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (M.J.S.-A.); (F.T.); (R.J.H.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| | - Michael D. Johnson
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| | - Yingratana A. McLennan
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| | - Ye Hyun Hwang
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (Y.H.H.); (F.T.)
| | - Pablo Juarez
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| | - Erin Lucille McBride
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| | - Adriana P. Pantoja
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, UC Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (M.J.S.-A.); (F.T.); (R.J.H.); (V.M.-C.)
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (Y.H.H.); (F.T.)
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (M.J.S.-A.); (F.T.); (R.J.H.); (V.M.-C.)
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA 95817, USA;
| | - Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA 95616, USA; (M.J.S.-A.); (F.T.); (R.J.H.); (V.M.-C.)
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA; (M.J.S.-A.); (M.D.J.); (Y.A.M.); (P.J.); (E.L.M.); (A.P.P.); (V.M.-C.)
| |
Collapse
|
24
|
Aishworiya R, Chi MH, Zafarullah M, Mendoza G, Ponzini MD, Kim K, Biag HMB, Thurman AJ, Abbeduto L, Hessl D, Randol JL, Bolduc FV, Jacquemont S, Lippé S, Hagerman P, Hagerman R, Schneider A, Tassone F. Intercorrelation of Molecular Biomarkers and Clinical Phenotype Measures in Fragile X Syndrome. Cells 2023; 12:1920. [PMID: 37508583 PMCID: PMC10377864 DOI: 10.3390/cells12141920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
This study contributes to a greater understanding of the utility of molecular biomarkers to identify clinical phenotypes of fragile X syndrome (FXS). Correlations of baseline clinical trial data (molecular measures-FMR1 mRNA, CYFIP1 mRNA, MMP9 and FMRP protein expression levels, nonverbal IQ, body mass index and weight, language level, NIH Toolbox, adaptive behavior rating, autism, and other mental health correlates) of 59 participants with FXS ages of 6-32 years are reported. FMR1 mRNA expression levels correlated positively with adaptive functioning levels, expressive language, and specific NIH Toolbox measures. The findings of a positive correlation of MMP-9 levels with obesity, CYFIP1 mRNA with mood and autistic symptoms, and FMR1 mRNA expression level with better cognitive, language, and adaptive functions indicate potential biomarkers for specific FXS phenotypes. These may be potential markers for future clinical trials for targeted treatments of FXS.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore 119074, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Mei-Hung Chi
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Psychiatry, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA (G.M.)
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA (G.M.)
| | - Matthew Dominic Ponzini
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Public Health Sciences, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Kyoungmi Kim
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Public Health Sciences, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Hazel Maridith Barlahan Biag
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Angela John Thurman
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Leonard Abbeduto
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Jamie Leah Randol
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA (G.M.)
- Integrative Genetics and Genomics Graduate Group, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
- UC Davis Biotechnology Program, University of California Davis, Davis, CA 95616, USA
| | - Francois V. Bolduc
- Department of Pediatrics, Department of Medical Genetics, Women and Children Health Research Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sebastien Jacquemont
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sarah Lippé
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Psychology, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Paul Hagerman
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA (G.M.)
| | - Randi Hagerman
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Andrea Schneider
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Flora Tassone
- MIND Institute, University of California Davis Medical Center, Sacramento, CA 95817, USA; (R.A.); (M.D.P.); (H.M.B.B.)
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA (G.M.)
| |
Collapse
|
25
|
da Silva CP, Camuzi D, Reis AHDO, Gonçalves AP, Dos Santos JM, Machado FB, Medina-Acosta E, Soares-Lima SC, Santos-Rebouças CB. Identification of a novel epigenetic marker for typical and mosaic presentations of Fragile X syndrome. Expert Rev Mol Diagn 2023; 23:1273-1281. [PMID: 37970883 DOI: 10.1080/14737159.2023.2284782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Fragile X syndrome (FXS) is primarily due to CGG repeat expansions in the FMR1 gene. FMR1 alleles are classified as normal (N), intermediate (I), premutation (PM), and full mutation (FM). FXS patients often carry an FM, but size mosaicism can occur. Additionally, loss of methylation boundary upstream of repeats results in de novo methylation spreading to FMR1 promoter in FXS patients. RESEARCH DESIGN AND METHODS This pilot study investigated the methylation boundary and adjacent regions in 66 males with typical and atypical FXS aged 1 to 30 years (10.86 ± 6.48 years). AmplideX FMR1 mPCR kit was used to discriminate allele profiles and methylation levels. CpG sites were assessed by pyrosequencing. RESULTS 40 out of 66 FXS patients (60.6%) showed an exclusive FM (n = 40), whereas the remaining (n = 26) exhibited size mosaicism [10 PM_FM (15.15%); 10 N_FM (15.15%); 2 N_PM_FM (3%)]. Four patients (6.1%) had deletions near repeats. Noteworthy, a CpG within FMR1 intron 2 displayed hypomethylation in FXS patients and hypermethylation in controls, demonstrating remarkable specificity, sensitivity, and accuracy when a methylation threshold of 69.5% was applied. CONCLUSIONS Since intragenic methylation is pivotal in gene regulation, the intronic CpG might be a novel epigenetic biomarker for FXS diagnosis.
Collapse
Affiliation(s)
- Camilla Pereira da Silva
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego Camuzi
- Molecular Carcinogenesis Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Adriana Helena de Oliveira Reis
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andressa Pereira Gonçalves
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jussara Mendonça Dos Santos
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Filipe Brum Machado
- Department of Biological Sciences, Minas Gerais State University, Minas Gerais, Brazil
| | - Enrique Medina-Acosta
- Biotechnology Laboratory, Molecular Diagnostic, and Research Center, State University of the North Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brazil
| | | | - Cíntia Barros Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Protic D, Polli R, Hwang YH, Mendoza G, Hagerman R, Durbin-Johnson B, Hayward BE, Usdin K, Murgia A, Tassone F. Activation Ratio Correlates with IQ in Female Carriers of the FMR1 Premutation. Cells 2023; 12:1711. [PMID: 37443745 PMCID: PMC10341054 DOI: 10.3390/cells12131711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Carriers of the FMR1 premutation (PM) allele are at risk of one or more clinical conditions referred to as FX premutation-associated conditions (FXPAC). Since the FMR1 gene is on the X chromosome, the activation ratio (AR) may impact the risk, age of onset, progression, and severity of these conditions. The aim of this study was to evaluate the reliability of AR measured using different approaches and to investigate potential correlations with clinical outcomes. Molecular and clinical assessments were obtained for 30 PM female participants, and AR was assessed using both Southern blot analysis (AR-Sb) and methylation PCR (AR-mPCR). Higher ARs were associated with lower FMR1 transcript levels for any given repeat length. The higher AR-Sb was significantly associated with performance, verbal, and full-scale IQ scores, confirming previous reports. However, the AR-mPCR was not significantly associated (p > 0.05) with these measures. Similarly, the odds of depression and the number of medical conditions were correlated with higher AR-Sb but not correlated with a higher AR-mPCR. This study suggests that AR-Sb may be a more reliable measure of the AR in female carriers of PM alleles. However, further studies are warranted in a larger sample size to fully evaluate the methylation status in these participants and how it may affect the clinical phenotype.
Collapse
Affiliation(s)
- Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Roberta Polli
- Laboratory of Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, 35128 Padova, Italy; (R.P.); (A.M.)
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35128 Padova, Italy
| | - Ye Hyun Hwang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (Y.H.H.); (G.M.)
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (Y.H.H.); (G.M.)
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, Sacramento, CA 95817, USA;
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Blythe Durbin-Johnson
- Department of Public Health Sciences, Division of Biostatistics, University of California, Davis, CA 95616, USA;
| | - Bruce E. Hayward
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.E.H.); (K.U.)
| | - Karen Usdin
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.E.H.); (K.U.)
| | - Alessandra Murgia
- Laboratory of Molecular Genetics of Neurodevelopment, Department of Woman and Child Health, University of Padova, 35128 Padova, Italy; (R.P.); (A.M.)
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35128 Padova, Italy
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (Y.H.H.); (G.M.)
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, Sacramento, CA 95817, USA;
| |
Collapse
|
27
|
Ciobanu CG, Nucă I, Popescu R, Antoci LM, Caba L, Ivanov AV, Cojocaru KA, Rusu C, Mihai CT, Pânzaru MC. Narrative Review: Update on the Molecular Diagnosis of Fragile X Syndrome. Int J Mol Sci 2023; 24:9206. [PMID: 37298158 PMCID: PMC10252420 DOI: 10.3390/ijms24119206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/30/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
The diagnosis and management of fragile X syndrome (FXS) have significantly improved in the last three decades, although the current diagnostic techniques are not yet able to precisely identify the number of repeats, methylation status, level of mosaicism, and/or the presence of AGG interruptions. A high number of repeats (>200) in the fragile X messenger ribonucleoprotein 1 gene (FMR1) results in hypermethylation of promoter and gene silencing. The actual molecular diagnosis is performed using a Southern blot, TP-PCR (Triplet-Repeat PCR), MS-PCR (Methylation-Specific PCR), and MS-MLPA (Methylation-Specific MLPA) with some limitations, with multiple assays being necessary to completely characterise a patient with FXS. The actual gold standard diagnosis uses Southern blot; however, it cannot accurately characterise all cases. Optical genome mapping is a new technology that has also been developed to approach the diagnosis of fragile X syndrome. Long-range sequencing represented by PacBio and Oxford Nanopore has the potential to replace the actual diagnosis and offers a complete characterization of molecular profiles in a single test. The new technologies have improved the diagnosis of fragile X syndrome and revealed unknown aberrations, but they are a long way from being used routinely in clinical practice.
Collapse
Affiliation(s)
- Cristian-Gabriel Ciobanu
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
| | - Irina Nucă
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
- Investigatii Medicale Praxis, St. Moara de Vant No 35, 700376 Iasi, Romania
| | - Roxana Popescu
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
- Medical Genetics Department, “Saint Mary” Emergency Children’s Hospital, St. Vasile Lupu No 62, 700309 Iasi, Romania
| | - Lucian-Mihai Antoci
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
| | - Lavinia Caba
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
| | - Anca Viorica Ivanov
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania
| | - Karina-Alexandra Cojocaru
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania
| | - Cristina Rusu
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
- Medical Genetics Department, “Saint Mary” Emergency Children’s Hospital, St. Vasile Lupu No 62, 700309 Iasi, Romania
| | | | - Monica-Cristina Pânzaru
- Medical Genetics Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No 16, 700115 Iasi, Romania; (C.-G.C.)
- Medical Genetics Department, “Saint Mary” Emergency Children’s Hospital, St. Vasile Lupu No 62, 700309 Iasi, Romania
| |
Collapse
|
28
|
Aishworiya R, Hwang YH, Santos E, Hayward B, Usdin K, Durbin-Johnson B, Hagerman R, Tassone F. Clinical implications of somatic allele expansion in female FMR1 premutation carriers. Sci Rep 2023; 13:7050. [PMID: 37120588 PMCID: PMC10148869 DOI: 10.1038/s41598-023-33528-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/14/2023] [Indexed: 05/01/2023] Open
Abstract
Carriers of a premutation allele (PM) in the FMR1 gene are at risk of developing a number of Fragile X premutation asssociated disorders (FXPAC), including Fragile X-associated Tremor/Ataxia Syndrome (FXTAS), Fragile X-associated Primary Ovarian Insufficiency (FXPOI), and Fragile X-associated neuropsychiatric disorders (FXAND). We have recently reported somatic CGG allele expansion in female PM; however, its clinical significance remains unclear. The aim of this study was to examine the potential clinical association between somatic FMR1 allele instability and PM associated disorders. Participants comprised of 424 female PM carriers age 0.3- 90 years. FMR1 molecular measures and clinical information on the presence of medical conditions, were determined for all subjects for primary analysis. Two sub-groups of participants (age ≥ 25, N = 377 and age ≥ 50, N = 134) were used in the analysis related to presence of FXPOI and FXTAS, respectively. Among all participants (N = 424), the degree of instability (expansion) was significantly higher (median 2.5 vs 2.0, P = 0.026) in participants with a diagnosis of attention deficit hyperactivity disorder (ADHD) compared to those without. FMR1 mRNA expression was significantly higher in subjects with any psychiatric disorder diagnosis (P = 0.0017); specifically, in those with ADHD (P = 0.009), and with depression (P = 0.025). Somatic FMR1 expansion was associated with the presence of ADHD in female PM and FMR1 mRNA levels were associated with the presence of mental health disorders. The findings of our research are innovative as they suggest a potential role of the CGG expansion in the clinical phenotype of PM and may potentially guide clinical prognosis and management.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50Th Street, Sacramento, CA, 95817, USA
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Singapore
| | - Ye Hyun Hwang
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, 4610 X St, Sacramento, CA, 95817, USA
| | - Ellery Santos
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50Th Street, Sacramento, CA, 95817, USA
- Department of Pediatrics, University of California Davis, School of Medicine, 4610 X St, Sacramento, CA, 95817, USA
| | - Bruce Hayward
- Laboratory of Cell and Molecular Biology, Digestive and Kidney Diseases, National Institute of Diabetes, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Karen Usdin
- Laboratory of Cell and Molecular Biology, Digestive and Kidney Diseases, National Institute of Diabetes, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Blythe Durbin-Johnson
- Department of Public Health Sciences, University of California, Davis, School of Medicine, 4610 X St, Sacramento, CA, 95817, USA
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50Th Street, Sacramento, CA, 95817, USA
- Department of Pediatrics, University of California Davis, School of Medicine, 4610 X St, Sacramento, CA, 95817, USA
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50Th Street, Sacramento, CA, 95817, USA.
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, 4610 X St, Sacramento, CA, 95817, USA.
| |
Collapse
|
29
|
Zafarullah M, Li J, Tseng E, Tassone F. Structure and Alternative Splicing of the Antisense FMR1 (ASFMR1) Gene. Mol Neurobiol 2023; 60:2051-2061. [PMID: 36598648 PMCID: PMC10461537 DOI: 10.1007/s12035-022-03176-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 12/10/2022] [Indexed: 01/05/2023]
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder caused by an expansion of 55-200 CGG repeats (premutation) in the 5'-UTR of the FMR1 gene. Bidirectional transcription at FMR1 locus has been demonstrated and specific alternative splicing of the Antisense FMR1 (ASFMR1) gene has been proposed to have a contributing role in the pathogenesis of FXTAS. The structure of ASFMR1 gene is still uncharacterized and it is currently unknown how many isoforms of the gene are expressed and at what level in premutation carriers (PM) and if they may contribute to the premutation pathology. In this study, we characterized the ASFMR1 gene structure and the transcriptional landscape by using PacBio SMRT sequencing with target enrichment (IDT customized probe panel). We identified 45 ASFMR1 isoforms ranging in sizes from 523 bp to 6 Kb, spanning approximately 59 kb of genomic DNA. Multiplexing and sequencing of six human brain samples from PM samples and normal control (HC) were carried out on the PacBio Sequel platform. We validated the presence of these isoforms by qRT-PCR and Sanger sequencing and characterized the acceptor and donor splicing site consensus sequences. Consistent with previous studies conducted in other tissue types, we found a high expression of ASFMR1 isoform Iso131bp in brain samples of PM as compared to HC, while no differences in expression levels were observed for the newly identified isoforms IsoAS1 and IsoAS2. We investigated the role of the splicing regulatory protein Sam68 which we did not observe in the alternative splicing of the ASFMR1 gene. Our study provides a useful insight into the structure of ASFMR1 gene and transcriptional landscape along with the expression pattern of various newly identified novel isoforms and on their potential role in premutation pathology.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Jie Li
- Bioinformatics Core, Genome Center, University of California Davis, Davis, CA, 95616, USA
| | | | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA.
- MIND Institute, University of California Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
30
|
Jam HZ, Li Y, DeVito R, Mousavi N, Ma N, Lujumba I, Adam Y, Maksimov M, Huang B, Dolzhenko E, Qiu Y, Kakembo FE, Joseph H, Onyido B, Adeyemi J, Bakhtiari M, Park J, Javadzadeh S, Jjingo D, Adebiyi E, Bafna V, Gymrek M. A deep population reference panel of tandem repeat variation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531600. [PMID: 36945429 PMCID: PMC10028971 DOI: 10.1101/2023.03.09.531600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Tandem repeats (TRs) represent one of the largest sources of genetic variation in humans and are implicated in a range of phenotypes. Here we present a deep characterization of TR variation based on high coverage whole genome sequencing from 3,550 diverse individuals from the 1000 Genomes Project and H3Africa cohorts. We develop a method, EnsembleTR, to integrate genotypes from four separate methods resulting in high-quality genotypes at more than 1.7 million TR loci. Our catalog reveals novel sequence features influencing TR heterozygosity, identifies population-specific trinucleotide expansions, and finds hundreds of novel eQTL signals. Finally, we generate a phased haplotype panel which can be used to impute most TRs from nearby single nucleotide polymorphisms (SNPs) with high accuracy. Overall, the TR genotypes and reference haplotype panel generated here will serve as valuable resources for future genome-wide and population-wide studies of TRs and their role in human phenotypes.
Collapse
Affiliation(s)
- Helyaneh Ziaei Jam
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Yang Li
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Ross DeVito
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Nima Mousavi
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA
| | - Nichole Ma
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Ibra Lujumba
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala-Uganda
| | - Yagoub Adam
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Mikhail Maksimov
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Bonnie Huang
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | | | - Yunjiang Qiu
- Illumina Incorporated, San Diego, California 92122, USA
| | - Fredrick Elishama Kakembo
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala-Uganda
| | - Habi Joseph
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala-Uganda
| | - Blessing Onyido
- Department of Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Jumoke Adeyemi
- Department of Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Mehrdad Bakhtiari
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Jonghun Park
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Sara Javadzadeh
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Daudi Jjingo
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, the Infectious Diseases Institute, Makerere University, Kampala-Uganda
- Department of Computer Science, Makerere University, Kampala, Uganda
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun, 112233, Nigeria
- Department of Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun, 112233, Nigeria
- Applied Bioinformatics Division, German Cancer Research Center (DKFZ), Heidelberg, Baden-Württemberg, 69120, Germany
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
| | - Melissa Gymrek
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
31
|
Ranjan R, Jha S, Prajjwal P, Chaudhary A, Dudeja P, Vora N, Mateen MA, Yousuf MA, Chaudhary B. Neurological, Psychiatric, and Multisystemic Involvement of Fragile X Syndrome Along With Its Pathophysiology, Methods of Screening, and Current Treatment Modalities. Cureus 2023; 15:e35505. [PMID: 37007359 PMCID: PMC10050793 DOI: 10.7759/cureus.35505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2023] [Indexed: 03/01/2023] Open
Abstract
Fragile X syndrome (FXS) is a hereditary disease that predominantly leads to intellectual disability (ID) in boys. It is the second prominent cause of ID, which manifests as a result of the atypical development of the cytosine-guanine-guanine (CGG) region. This irregular extension of the CGG region gives rise to methylation and silencing of the fragile X mental retardation 1 (FMR1) gene, causing a loss of the fragile X mental retardation 1 protein (FMRP). This reduction or loss of FMRP is the main cause of ID. It has a multisystemic involvement showing neuropsychiatric features such as ID, speech and language delay, autism spectrum disorder, sensory hyperarousal, social anxiety, abnormal eye contact, shyness, and aggressive behaviour. It is also known to cause musculoskeletal symptoms, ocular symptoms, cardiac abnormalities, and gastrointestinal symptoms. The management is challenging, and there is no known cure for the disease; hence an early diagnosis of the condition is needed through prenatal screening offered to couples with familial history of ID before conception. The management rests on non-pharmacological modalities, including applied behaviour analysis, physical therapy, occupational therapy, speech-language therapy, and pharmacologic management through symptomatic treatment of comorbid behaviours and psychiatric problems and some forms of targeted therapy.
Collapse
|
32
|
Aishworiya R, Protic D, Tang SJ, Schneider A, Tassone F, Hagerman R. Fragile X-Associated Neuropsychiatric Disorders (FXAND) in Young Fragile X Premutation Carriers. Genes (Basel) 2022; 13:genes13122399. [PMID: 36553666 PMCID: PMC9778214 DOI: 10.3390/genes13122399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Background: The fragile X premutation carrier state (PM) (55-200 CGG repeats in the fragile X messenger ribonucleoprotein 1, FMR1 gene) is associated with several conditions, including fragile X-associated primary ovarian insufficiency (FXPOI) and fragile X-associated tremor ataxia (FXTAS), with current literature largely primarily investigating older PM individuals. The aim of this study was to identify the prevalence of fragile X-associated neurodevelopmental disorders (FXAND) in a sample of young PM individuals. Methods: This was a retrospective study conducted through a medical record review of PM individuals who were seen either for clinical concerns (probands, 45.9%) or identified through the cascade testing (non-probands, 54.1%) of an affected sibling with fragile X syndrome. Information on the presence of autism spectrum disorder, attention deficit hyperactivity disorder, anxiety, depression, long-term psychiatric medication intake, and cognitive function, based on standardized assessments, was obtained. Molecular data, including CGG repeat number and FMR1 mRNA levels, were also available for a subset of participants. Analysis included descriptive statistics and a test of comparison to describe the clinical profile of PM individuals pertinent to FXAND. Results: Participants included 61 individuals (52 males and 9 females) aged 7.8 to 20.0 years (mean 12.6 ± 3.4) with a mean full-scale IQ of 90.9 ± 22.7. The majority (N = 52; 85.2%) had at least one mental health disorder, with anxiety being the most common (82.0% of subjects), followed by ADHD (66.5%), and ASD (32.8%). Twenty-seven (87.1%) of non-probands also had at least one mental health condition, with probands having lower cognitive and adaptive skills than non-probands. ASD was present in 20 participants (17/52 males and 3/9 females; 15 probands) with significantly lower FSIQ in those with ASD (mean 73.5 vs. 98.0, p < 0.001). Participants with ASD had a higher number of long-term medications compared to those without (2.32 vs. 1.3, p = 0.002). Conclusions: Our findings indicate a high rate of FXAND diagnoses within a cohort of young PM individuals, including those identified via cascade testing, although this was not a population sample. An awareness of the entity of FXAND and the early recognition of the symptoms of associated conditions may facilitate timely and appropriate care for PM individuals.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, Singapore
- Correspondence: ; Tel.: +916-703-0247; Fax: +916-703-0240
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Si Jie Tang
- Department of Pediatrics, School of Medicine, University of California Davis, 4610 X St, Sacramento, CA 95817, USA
| | - Andrea Schneider
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Pediatrics, School of Medicine, University of California Davis, 4610 X St, Sacramento, CA 95817, USA
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, 4610 X St, Sacramento, CA 95817, USA
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Pediatrics, School of Medicine, University of California Davis, 4610 X St, Sacramento, CA 95817, USA
| |
Collapse
|
33
|
Berry-Kravis E, Hagerman R, Budimirovic D, Erickson C, Heussler H, Tartaglia N, Cohen J, Tassone F, Dobbins T, Merikle E, Sebree T, Tich N, Palumbo JM, O’Quinn S. A randomized, controlled trial of ZYN002 cannabidiol transdermal gel in children and adolescents with fragile X syndrome (CONNECT-FX). J Neurodev Disord 2022; 14:56. [PMID: 36434514 PMCID: PMC9700889 DOI: 10.1186/s11689-022-09466-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 11/03/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is associated with dysregulated endocannabinoid signaling and may therefore respond to cannabidiol therapy. DESIGN CONNECT-FX was a double-blind, randomized phase 3 trial assessing efficacy and safety of ZYN002, transdermal cannabidiol gel, for the treatment of behavioral symptoms in children and adolescents with FXS. METHODS Patients were randomized to 12 weeks of ZYN002 (250 mg or 500 mg daily [weight-based]) or placebo, as add-on to standard of care. The primary endpoint assessed change in social avoidance (SA) measured by the Aberrant Behavior Checklist-Community Edition FXS (ABC-CFXS) SA subscale in a full cohort of patients with a FXS full mutation, regardless of the FMR1 methylation status. Ad hoc analyses assessed efficacy in patients with ≥ 90% and 100% methylation of the promoter region of the FMR1 gene, in whom FMR1 gene silencing is most likely. RESULTS A total of 212 patients, mean age 9.7 years, 75% males, were enrolled. A total of 169 (79.7%) patients presented with ≥ 90% methylation of the FMR1 promoter and full mutation of FMR1. Although statistical significance for the primary endpoint was not achieved in the full cohort, significant improvement was demonstrated in patients with ≥ 90% methylation of FMR1 (nominal P = 0.020). This group also achieved statistically significant improvements in Caregiver Global Impression-Change in SA and isolation, irritable and disruptive behaviors, and social interactions (nominal P-values: P = 0.038, P = 0.028, and P = 0.002). Similar results were seen in patients with 100% methylation of FMR1. ZYN002 was safe and well tolerated. All treatment-emergent adverse events (TEAEs) were mild or moderate. The most common treatment-related TEAE was application site pain (ZYN002: 6.4%; placebo: 1.0%). CONCLUSIONS In CONNECT-FX, ZYN002 was well tolerated in patients with FXS and demonstrated evidence of efficacy with a favorable benefit risk relationship in patients with ≥ 90% methylation of the FMR1 gene, in whom gene silencing is most likely, and the impact of FXS is typically most severe. TRIAL REGISTRATION The CONNECT-FX trial is registered on Clinicaltrials.gov (NCT03614663).
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- grid.240684.c0000 0001 0705 3621Departments of Pediatrics and Neurological Sciences, Rush University Medical Center, Chicago, IL USA
| | - Randi Hagerman
- grid.413079.80000 0000 9752 8549Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California-Davis Medical Center, Sacramento, CA USA ,grid.27860.3b0000 0004 1936 9684Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA USA
| | - Dejan Budimirovic
- grid.21107.350000 0001 2171 9311Departments of Psychiatry and Child Psychiatry, Fragile X Clinic, Kennedy Krieger Institute/the Johns Hopkins Medical Institutions, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Craig Erickson
- grid.24827.3b0000 0001 2179 9593Department of Psychiatry and Behavioral Neuroscience, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Helen Heussler
- grid.512914.a0000 0004 0642 3960Centre for Clinical Trials in Rare Neurodevelopmental Disorders, Children’s Health Queensland, Brisbane, Australia ,grid.1003.20000 0000 9320 7537Centre for Child Health Research, University of Queensland, Brisbane, Australia
| | - Nicole Tartaglia
- Department of Pediatrics, Developmental Pediatrics, University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO USA
| | - Jonathan Cohen
- Fragile X Alliance Inc, North Caulfield, VIC, Australia ,grid.1002.30000 0004 1936 7857Centre for Developmental Disability Health Victoria, Monash University, Clayton, VIC Australia
| | - Flora Tassone
- grid.413079.80000 0000 9752 8549Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California-Davis Medical Center, Sacramento, CA USA ,grid.413079.80000 0000 9752 8549Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA USA
| | | | | | - Terri Sebree
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| | - Nancy Tich
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| | - Joseph M. Palumbo
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| | - Stephen O’Quinn
- grid.422480.80000 0004 8307 0679Zynerba Pharmaceuticals Inc., Devon, PA USA
| |
Collapse
|
34
|
Hocking DR, Loesch DZ, Stimpson P, Tassone F, Atkinson A, Storey E. Relationships of Motor Changes with Cognitive and Neuropsychiatric Features in FMR1 Male Carriers Affected with Fragile X-Associated Tremor/Ataxia Syndrome. Brain Sci 2022; 12:brainsci12111549. [PMID: 36421873 PMCID: PMC9688438 DOI: 10.3390/brainsci12111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The premutation expansion of the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene on the X chromosome has been linked to a range of clinical and subclinical features. Nearly half of men with FMR1 premutation develop a neurodegenerative disorder; Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). In this syndrome, cognitive executive decline and psychiatric changes may co-occur with major motor features, and in this study, we explored the interrelationships between these three domains in a sample of adult males affected with FXTAS. A sample of 23 adult males aged between 48 and 80 years (mean = 62.3; SD = 8.8), carrying premutation expansions between 45 and 118 CGG repeats, and affected with FXTAS, were included in this study. We employed a battery of cognitive assessments, two standard motor rating scales, and two self-reported measures of psychiatric symptoms. When controlling for age and/or educational level, where appropriate, there were highly significant correlations between motor rating score for ICARS gait domain, and the scores representing global cognitive decline (ACE-III), processing speed (SDMT), immediate memory (Digit Span), and depression and anxiety scores derived from both SCL90 and DASS instruments. Remarkably, close relationships of UPDRS scores, representing the contribution of Parkinsonism to FXTAS phenotypes, were exclusive to psychiatric scores. Highly significant relationships between CGG repeat size and most scores for three phenotypic domains suggest a close tracking with genetic liability. These findings of relationships between a constellation of phenotypic domains in male PM carriers with FXTAS are reminiscent of other conditions associated with disruption to cerebro-cerebellar circuits.
Collapse
Affiliation(s)
- Darren R. Hocking
- Developmental Neuromotor & Cognition Lab, School of Psychology and Public Health, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence:
| | - Danuta Z. Loesch
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC 3086, Australia
| | - Paige Stimpson
- Psychology Department, Monash Health, Clayton, VIC 3068, Australia
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, M.I.N.D. Institute, School of Medicine, University of California Davis Medical Center, University of California, Davis, Davis, CA 95616, USA
| | - Anna Atkinson
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC 3086, Australia
| | - Elsdon Storey
- Department of Medicine (Neuroscience), Alfred Hospital Campus, Monash University, Melbourne, VIC 3068, Australia
| |
Collapse
|
35
|
Liang Q, Liu Y, Liu Y, Duan R, Meng W, Zhan J, Xia J, Mao A, Liang D, Wu L. Comprehensive Analysis of Fragile X Syndrome: Full Characterization of the FMR1 Locus by Long-Read Sequencing. Clin Chem 2022; 68:1529-1540. [PMID: 36171182 DOI: 10.1093/clinchem/hvac154] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/21/2022] [Indexed: 11/14/2022]
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most frequent cause of inherited X-linked intellectual disability. Conventional FXS genetic testing methods mainly focus on FMR1 CGG expansions and fail to identify AGG interruptions, rare intragenic variants, and large gene deletions. METHODS A long-range PCR and long-read sequencing-based assay termed comprehensive analysis of FXS (CAFXS) was developed and evaluated in Coriell and clinical samples by comparing to Southern blot analysis and triplet repeat-primed PCR (TP-PCR). RESULTS CAFXS accurately detected the number of CGG repeats in the range of 93 to at least 940 with mass fraction of 0.5% to 1% in the background of normal alleles, which was 2-4-fold analytically more sensitive than TP-PCR. All categories of mutations detected by control methods, including full mutations in 30 samples, were identified by CAFXS for all 62 clinical samples. CAFXS accurately determined AGG interruptions in all 133 alleles identified, even in mosaic alleles. CAFXS successfully identified 2 rare intragenic variants including the c.879A > C variant in exon 9 and a 697-bp microdeletion flanking upstream of CGG repeats, which disrupted primer annealing in TP-PCR assay. In addition, CAFXS directly determined the breakpoints of a 237.1-kb deletion and a 774.0-kb deletion encompassing the entire FMR1 gene in 2 samples. CONCLUSIONS Long-read sequencing-based CAFXS represents a comprehensive assay for identifying FMR1 CGG expansions, AGG interruptions, rare intragenic variants, and large gene deletions, which greatly improves the genetic screening and diagnosis for FXS.
Collapse
Affiliation(s)
- Qiaowei Liang
- Department of Medical Genetics, Hunan Jiahui Genetics Hospital, Changsha, Hunan, China
| | - Yingdi Liu
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yaning Liu
- Department of Medical Genetics, Hunan Jiahui Genetics Hospital, Changsha, Hunan, China
| | - Ranhui Duan
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Wanli Meng
- Berry Genomics Corporation, Beijing, China
| | | | - Jiahui Xia
- Department of Medical Genetics, Hunan Jiahui Genetics Hospital, Changsha, Hunan, China
| | - Aiping Mao
- Berry Genomics Corporation, Beijing, China
| | - Desheng Liang
- Department of Medical Genetics, Hunan Jiahui Genetics Hospital, Changsha, Hunan, China.,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Lingqian Wu
- Department of Medical Genetics, Hunan Jiahui Genetics Hospital, Changsha, Hunan, China.,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
36
|
Saldarriaga W, González-Teshima LY, Forero-Forero JV, Tang HT, Tassone F. Mosaicism in Fragile X syndrome: A family case series. JOURNAL OF INTELLECTUAL DISABILITIES : JOID 2022; 26:800-807. [PMID: 33998336 DOI: 10.1177/1744629521995346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Fragile X syndrome (FXS) has a classic phenotype, however its expression can be variable among full mutation males. This is secondary to variable methylation mosaicisms and the number of CGG triplet repeats in the non-coding region of the Fragile X Mental Retardation 1 (FMR1) gene, producing a variable expression of the Fragile X Mental Retardation Protein (FMRP). Here we report a family with several individuals affected by FXS: a boy with a hypermethylated FMR1 mutation and a classic phenotype; a man with an FMR1 gene mosaicism in the range of premutation (PM) and full mutation (FM), who has a mild phenotype due to which FXS was initially disregarded; and the cases of four women with a FM and mosaicism. This report highlights the importance of DNA molecular testing for the diagnosis of FXS in patients with developmental delay, intellectual disability and/or autism due to the variable phenotype that occurs in individuals with FMR1 mosaicisms.
Collapse
|
37
|
Hwang YH, Hayward BE, Zafarullah M, Kumar J, Durbin Johnson B, Holmans P, Usdin K, Tassone F. Both cis and trans-acting genetic factors drive somatic instability in female carriers of the FMR1 premutation. Sci Rep 2022; 12:10419. [PMID: 35729184 PMCID: PMC9213438 DOI: 10.1038/s41598-022-14183-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/18/2022] [Indexed: 11/30/2022] Open
Abstract
The fragile X mental retardation (FMR1) gene contains an expansion-prone CGG repeat within its 5' UTR. Alleles with 55-200 repeats are known as premutation (PM) alleles and confer risk for one or more of the FMR1 premutation (PM) disorders that include Fragile X-associated Tremor/Ataxia Syndrome (FXTAS), Fragile X-associated Primary Ovarian Insufficiency (FXPOI), and Fragile X-Associated Neuropsychiatric Disorders (FXAND). PM alleles expand on intergenerational transmission, with the children of PM mothers being at risk of inheriting alleles with > 200 CGG repeats (full mutation FM) alleles) and thus developing Fragile X Syndrome (FXS). PM alleles can be somatically unstable. This can lead to individuals being mosaic for multiple size alleles. Here, we describe a detailed evaluation of somatic mosaicism in a large cohort of female PM carriers and show that 94% display some evidence of somatic instability with the presence of a series of expanded alleles that differ from the next allele by a single repeat unit. Using two different metrics for instability that we have developed, we show that, as with intergenerational instability, there is a direct relationship between the extent of somatic expansion and the number of CGG repeats in the originally inherited allele and an inverse relationship with the number of AGG interruptions. Expansions are progressive as evidenced by a positive correlation with age and by examination of blood samples from the same individual taken at different time points. Our data also suggests the existence of other genetic or environmental factors that affect the extent of somatic expansion. Importantly, the analysis of candidate single nucleotide polymorphisms (SNPs) suggests that two DNA repair factors, FAN1 and MSH3, may be modifiers of somatic expansion risk in the PM population as observed in other repeat expansion disorders.
Collapse
Affiliation(s)
- Ye Hyun Hwang
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Bruce Eliot Hayward
- Laboratory of Molecular and Cellular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Jay Kumar
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Blythe Durbin Johnson
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Peter Holmans
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | - Karen Usdin
- Laboratory of Molecular and Cellular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA, 95817, USA. .,MIND Institute, University of California Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
38
|
Jiraanont P, Manor E, Tabatadze N, Zafarullah M, Mendoza G, Melikishvili G, Tassone F. De Novo Large Deletion Leading to Fragile X Syndrome. Front Genet 2022; 13:884424. [PMID: 35646065 PMCID: PMC9130735 DOI: 10.3389/fgene.2022.884424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is the most frequent cause of X-linked inherited intellectual disabilities (ID) and the most frequent monogenic form of autism spectrum disorders. It is caused by an expansion of a CGG trinucleotide repeat located in the 5'UTR of the FMR1 gene, resulting in the absence of the fragile X mental retardation protein, FMRP. Other mechanisms such as deletions or point mutations of the FMR1 gene have been described and account for approximately 1% of individuals with FXS. Here, we report a 7-year-old boy with FXS with a de novo deletion of approximately 1.1 Mb encompassing several genes, including the FMR1 and the ASFMR1 genes, and several miRNAs, whose lack of function could result in the observed proband phenotypes. In addition, we also demonstrate that FMR4 completely overlaps with ASFMR1, and there are no sequencing differences between both transcripts (i.e., ASFMR1/FMR4 throughout the article).
Collapse
Affiliation(s)
- Poonnada Jiraanont
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Esther Manor
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel.,Genetics Institute, Soroka Medical Center, Beersheba, Israel
| | - Nazi Tabatadze
- Department of Pediatrics, MediClub Georgia Medical Center, Tbilisi, Georgia
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Gia Melikishvili
- Department of Pediatrics, MediClub Georgia Medical Center, Tbilisi, Georgia
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States.,UC Davis MIND Institute, UC Davis Health, Sacramento, CA, United States
| |
Collapse
|
39
|
McLennan YA, Mosconi MW, McKenzie FJ, Famula J, Krawchuk B, Kim K, Clark CJ, Hessl D, Rivera SM, Simon TJ, Tassone F, Hagerman RJ. Prosaccade and Antisaccade Behavior in Fragile X-Associated Tremor/Ataxia Syndrome Progression. Mov Disord Clin Pract 2022; 9:473-478. [PMID: 35586536 PMCID: PMC9092736 DOI: 10.1002/mdc3.13449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 11/08/2022] Open
Abstract
Background Quantitative measurement of eye movements can reveal subtle progression in neurodegenerative diseases. Objective To determine if quantitative measurements of eye movements may reveal subtle progression of fragile X-associated tremor and ataxia (FXTAS). Methods Prosaccade (PS) and antisaccade (AS) behavior was analyzed in 25 controls, 57 non-FXTAS carriers, and 46 carriers with FXTAS. Results Symptomatic individuals with FXTAS had longer AS latencies, increased rates of AS errors, and increased AS dysmetria relative to non-FXTAS carriers and controls. These deficits, along with PS latency and velocity, were greater in advanced FXTAS stages. Conclusion AS deficits differentiated FXTAS from non-FXTAS premutation carriers implicating top-down control and frontostriatal deterioration. However, the absence of group differences between non-FXTAS carriers and controls in AS and PS markers suggests saccade performance may not be a sensitive enough measure for detecting conversion to FXTAS, but instead more helpful as translational biomarkers of FXTAS progression.
Collapse
Affiliation(s)
- Yingratana A. McLennan
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- Department of PediatricsUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
| | - Matthew W. Mosconi
- Life Span Institute, Kansas Center for Autism Research and Training, and Clinical Child Psychology ProgramUniversity of KansasLawrenceKansasUSA
| | | | - Jessica Famula
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
| | - Bennet Krawchuk
- University of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Kyoungmi Kim
- Department of PsychologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Courtney J. Clark
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- Department of PediatricsUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
| | - David Hessl
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- University of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Susan M. Rivera
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- Department of PsychologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Tony J. Simon
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- University of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Flora Tassone
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- Department of Biochemistry and Molecular MedicineUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Randi J. Hagerman
- The MIND InstituteUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
- Department of PediatricsUniversity of California Davis Medical CenterSacramentoCaliforniaUSA
| |
Collapse
|
40
|
Famula J, Ferrer E, Hagerman RJ, Tassone F, Schneider A, Rivera SM, Hessl D. Neuropsychological changes in FMR1 premutation carriers and onset of fragile X-associated tremor/ataxia syndrome. J Neurodev Disord 2022; 14:23. [PMID: 35321639 PMCID: PMC8942145 DOI: 10.1186/s11689-022-09436-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/14/2022] [Indexed: 11/24/2022] Open
Abstract
Background Carriers of the FMR1 premutation are at increased risk of developing a late-onset progressive neurodegenerative disease, fragile X-associated tremor/ataxia syndrome (FXTAS), characterized by intention tremor, gait ataxia, and cognitive decline. Cross-sectional studies to date have provided evidence that neuropsychological changes, such as executive function alterations, or subtle motor changes, may precede the onset of formal FXTAS, perhaps characterizing a prodromal state. However, the lack of longitudinal data has prevented the field from forming a clear picture of progression over time within individuals, and we lack consensus regarding early markers of risk and measures that may be used to track response to intervention. Methods This was a longitudinal study of 64 male FMR1 premutation carriers (Pm) without FXTAS at study entry and 30 normal controls (Nc), aged 40 to 80 years (Pm M = 60.0 years; Nc M = 57.4 years). Fifty of the Pm and 22 of the Nc were re-assessed after an average of 2.33 years, and 37 Pm and 20 Nc were re-assessed a third time after an average of another 2.15 years. Eighteen of 64 carriers (28%) converted to FXTAS during the study to date. Neuropsychological assessments at each time point, including components of the Cambridge Neuropsychological Test Automated Battery (CANTAB), tapped domains of episodic and working memory, inhibitory control, visual attention, planning, executive control of movement, and manual speed and dexterity. Age-based mixed models were used to examine group differences in change over time on the outcomes in the full sample, and differences were further evaluated in 15 trios (n = 45; 15 Pm “converters,” 15 Pm “nonconverters,” 15 Nc) that were one-one matched on age, education, and socioeconomic status. Results Compared to Nc, Pm showed significantly greater rates of change over time in visual working memory, motor dexterity, inhibitory control, and manual movement speed. After multiple comparison correction, significant effects remained for motor dexterity. Worsening inhibitory control and slower manual movements were related to progression in FXTAS stage, but these effects became statistically non-significant after correcting for multiple comparisons. Higher FMR1 mRNA correlated with worsening manual reaction time but did not survive multiple comparisons and no other molecular measures correlated with neuropsychological changes. Finally, trio comparisons revealed greater rate of decline in planning and manual movement speed in Pm converters compared to Pm nonconverters. Conclusions Accelerated decline in executive function and subtle motor changes, likely mediated by frontocerebellar circuits, may precede, and then track with the emergence of formal FXTAS symptoms. Further research to develop and harmonize clinical assessment of FMR1 carriers across centers is needed to prepare for future prophylactic and treatment trials for this disorder.
Collapse
Affiliation(s)
- Jessica Famula
- MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Emilio Ferrer
- Department of Psychology, University of California Davis, Davis, CA, USA
| | - Randi J Hagerman
- MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Flora Tassone
- MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - Andrea Schneider
- MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Susan M Rivera
- MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Psychology, University of California Davis, Davis, CA, USA.,Center for Mind and Brain, University of California Davis, Davis, CA, USA
| | - David Hessl
- MIND Institute, University of California Davis Health, 2825 50th Street, Sacramento, CA, 95817, USA. .,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
41
|
Meng L, Kaufmann WE, Frye RE, Ong K, Kaminski JW, Velinov M, Berry-Kravis E. The association between mosaicism type and cognitive and behavioral functioning among males with fragile X syndrome. Am J Med Genet A 2022; 188:858-866. [PMID: 35148024 PMCID: PMC10948005 DOI: 10.1002/ajmg.a.62594] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022]
Abstract
Mosaicism in fragile X syndrome (FXS) refers to two different FMR1 allele variations: size mosaicism represents different numbers of CGG repeats between the two alleles, such that in addition to a full mutation allele there is an allele in the normal or premutation range of CGG repeats, while methylation mosaicism indicates whether a full-mutation allele is fully or partially methylated. The present study explored the association between mosaicism type and cognitive and behavioral functioning in a large sample of males 3 years and older (n = 487) with FXS, participating in the Fragile X Online Registry with Accessible Research Database. Participants with methylation mosaicism were less severely cognitively affected as indicated by a less severe intellectual disability rating, higher intelligence quotient and adaptive behavior score, and lower social impairment score. In contrast, the presence of size mosaicism was not significantly associated with better cognitive and behavioral outcomes than full mutation. Our findings suggest that methylation mosaicism is associated with better cognitive functioning and adaptive behavior and less social impairment. Further research could assess to what extent these cognitive and behavioral differences depend on molecular diagnostic methods and the impact of mosaicism on prognosis of individuals with FXS.
Collapse
Affiliation(s)
- Lu Meng
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia, USA
| | - Walter E. Kaufmann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Richard E. Frye
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, USA
| | - Katherine Ong
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia, USA
| | - Jennifer W. Kaminski
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia, USA
| | - Milen Velinov
- Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics and Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
42
|
Protic DD, Aishworiya R, Salcedo-Arellano MJ, Tang SJ, Milisavljevic J, Mitrovic F, Hagerman RJ, Budimirovic DB. Fragile X Syndrome: From Molecular Aspect to Clinical Treatment. Int J Mol Sci 2022; 23:1935. [PMID: 35216055 PMCID: PMC8875233 DOI: 10.3390/ijms23041935] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
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.
Collapse
Affiliation(s)
- Dragana D. Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia
| | - Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Si Jie Tang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
| | - Jelena Milisavljevic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Filip Mitrovic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
43
|
Wang JY, Grigsby J, Placido D, Wei H, Tassone F, Kim K, Hessl D, Rivera SM, Hagerman RJ. Clinical and Molecular Correlates of Abnormal Changes in the Cerebellum and Globus Pallidus in Fragile X Premutation. Front Neurol 2022; 13:797649. [PMID: 35211082 PMCID: PMC8863211 DOI: 10.3389/fneur.2022.797649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/12/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Fragile X premutation carriers (55-200 CGG triplets) may develop a progressive neurodegenerative disorder, fragile X-associated tremor/ataxia syndrome (FXTAS), after the age of 50. The neuroradiologic markers of FXTAS are hyperintense T2-signals in the middle cerebellar peduncle-the MCP sign. We recently noticed abnormal T2-signals in the globus pallidus in male premutation carriers and controls but the prevalence and clinical significance were unknown. METHODS We estimated the prevalence of the MCP sign and pallidal T2-abnormalities in 230 male premutation carriers and 144 controls (aged 8-86), and examined the associations with FXTAS symptoms, CGG repeat length, and iron content in the cerebellar dentate nucleus and globus pallidus. RESULTS Among participants aged ≥45 years (175 premutation carriers and 82 controls), MCP sign was observed only in premutation carriers (52 vs. 0%) whereas the prevalence of pallidal T2-abnormalities approached significance in premutation carriers compared with controls after age-adjustment (25.1 vs. 13.4%, p = 0.069). MCP sign was associated with impaired motor and executive functioning, and the additional presence of pallidal T2-abnormalities was associated with greater impaired executive functioning. Among premutation carriers, significant iron accumulation was observed in the dentate nucleus, and neither pallidal or MCP T2-abnormalities affected measures of the dentate nucleus. While the MCP sign was associated with CGG repeat length >75 and dentate nucleus volume correlated negatively with CGG repeat length, pallidal T2-abnormalities did not correlate with CGG repeat length. However, pallidal signal changes were associated with age-related accelerated iron depletion and variability and having both MCP and pallidal signs further increased iron variability in the globus pallidus. CONCLUSIONS Only the MCP sign, not pallidal abnormalities, revealed independent associations with motor and cognitive impairment; however, the occurrence of combined MCP and pallidal T2-abnormalities may present a risk for greater cognitive impairment and increased iron variability in the globus pallidus.
Collapse
Affiliation(s)
- Jun Yi Wang
- Center for Mind and Brain, University of California, Davis, Davis, CA, United States
| | - Jim Grigsby
- Departments of Psychology and Medicine, University of Colorado Denver, Denver, CO, United States
| | - Diego Placido
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Hongjiang Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute for Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, United States
| | - Kyoungmi Kim
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - David Hessl
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, United States
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Susan M. Rivera
- Center for Mind and Brain, University of California, Davis, Davis, CA, United States
- Departments of Psychology and Medicine, University of Colorado Denver, Denver, CO, United States
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, United States
| | - Randi J. Hagerman
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, United States
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
44
|
Martins AAS, Paiva GM, Matosinho CGR, Coser EM, Fonseca PADS, Haase VG, Carvalho MRS. Working memory and arithmetic impairments in children with FMR1 premutation and gray zone alleles. Dement Neuropsychol 2022; 16:105-114. [PMID: 35719251 PMCID: PMC9170264 DOI: 10.1590/1980-5764-dn-2021-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/03/2021] [Indexed: 11/22/2022] Open
Abstract
Expansive mutations in familial mental retardation 1 (FMR1) gene have been associated with different phenotypes. Full mutations are associated with intellectual disability and autism spectrum disorder; premutations are associated with math learning difficulties and working memory impairments. In gray zone, neuropsychological development has not yet been described. Objectives This study aimed to describe the frequency of FMR1 premutation and gray zone alleles in a school population sample representing a broad spectrum of variation in math achievement and detail school achievement and cognitive performance in the children identified with FMR1 premutation or gray zone alleles. Methods We described a two-phase study. In the first phase, 2,195 school-age children were screened for math achievement. In the second phase, 378 children with normal intelligence were neuropsychologically assessed and genotyped for FMR1. Of these, 121 children (61 girls) performed below percentile 25 in mathematics (MD group) and 257 children (146 girls) performed above percentile 25 (control group). Results Four pupils presented expanded alleles, one premutation and three gray zone alleles. The girl with the premutation and one boy with a gray zone allele presented impairments in working memory and arithmetic performance below percentile 6, compatible with the diagnosis of developmental dyscalculia. These children's difficulties were not associated with inaccuracy of nonsymbolic number representations or literacy impairments. Dyscalculia in these children seems to be associated mainly with working memory impairments. Conclusions FMR1 expansions in the gray zone may contribute to dyscalculia in otherwise healthy and normally intelligent children.
Collapse
Affiliation(s)
- Aline Aparecida Silva Martins
- Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Postgraduate Program em Genética, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Belo Horizonte MG, Brazil
| | - Giulia Moreira Paiva
- Universidade Federal de Minas Gerais, Faculdade de Filosofia e Ciências Humanas, Departamento de Psicologia, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Programa de Pós-Graduação em Neurociências, Belo Horizonte MG, Brazil
| | - Carolina Guimarães Ramos Matosinho
- Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Postgraduate Program em Genética, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Belo Horizonte MG, Brazil
| | - Elisângela Monteiro Coser
- Fundação Oswaldo Cruz, Instituto René Rachou, Departamento de Informática de Biossistemas e Genômica, Belo Horizonte MG, Brazil
| | - Pablo Augusto de Souza Fonseca
- Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Postgraduate Program em Genética, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Belo Horizonte MG, Brazil
| | - Vitor Geraldi Haase
- Universidade Federal de Minas Gerais, Faculdade de Filosofia e Ciências Humanas, Departamento de Psicologia, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Programa de Pós-Graduação em Neurociências, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Faculdade de Medicina, Postgraduate Program em Saúde da Criança e do Adolescente Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Faculdade de Filosofia e Ciências Humanas, Departamento de Psicologia, Postgraduate Program em Psicologia, Belo Horizonte MG, Brazil.,Instituto Nacional de Ciência e Tecnologia em Cognição, Comportamento e Ensino, São Carlos SP, Brazil
| | - Maria Raquel Santos Carvalho
- Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Postgraduate Program em Genética, Belo Horizonte MG, Brazil.,Universidade Federal de Minas Gerais, Intituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Belo Horizonte MG, Brazil
| |
Collapse
|
45
|
Protic D, Salcedo-Arellano MJ, Stojkovic M, Saldarriaga W, Ávila Vidal LA, Miller RM, Tabatadze N, Peric M, Hagerman R, Budimirovic DB. Raising Knowledge and Awareness of Fragile X Syndrome in Serbia, Georgia, and Colombia: A Model for Other Developing Countries? THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2021; 94:559-571. [PMID: 34970093 PMCID: PMC8686783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
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.
Collapse
Affiliation(s)
- Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of
Medicine, University of Belgrade, Belgrade, Serbia
| | - Maria Jimena Salcedo-Arellano
- Department of Pediatrics, University of California Davis School of Medicine,
Sacramento, CA, USA,Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS,
University of California Davis, Sacramento, CA, USA,Department of Pathology and Laboratory Medicine, University of California Davis
School of Medicine, Sacramento, CA, USA
| | - Maja Stojkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of
Medicine, University of Belgrade, Belgrade, Serbia
| | - Wilmar Saldarriaga
- Department of Morphology, Universidad del Valle School of Medicine, Cali,
Colombia,Department of Obstetrics and Gynecology, Hospital Universitario del Valle,
Evaristo Garcia, Cali, Colombia
| | | | | | - Nazi Tabatadze
- Fragile X Center, MediClubGeorgia Medical Center, Tbilisi, Georgia
| | | | - Randi Hagerman
- Department of Pediatrics, University of California Davis School of Medicine,
Sacramento, CA, USA,Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS,
University of California Davis, Sacramento, CA, USA
| | - Dejan B. Budimirovic
- Departments of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute,
Baltimore, MD, USA,Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns
Hopkins School of Medicine, Baltimore, MD, USA,To whom all correspondence should be addressed: Dejan Budimirovic, MD,
Department of Psychiatry, Psychiatry Mental Health Program, Outpatient Services, Kennedy
Krieger Institute, Baltimore, MD;
| |
Collapse
|
46
|
FMR1 allele frequencies in 51,000 newborns: a large-scale population study in China. World J Pediatr 2021; 17:653-658. [PMID: 34738199 DOI: 10.1007/s12519-021-00473-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/05/2021] [Indexed: 10/19/2022]
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.
Collapse
|
47
|
FMRP Levels in Human Peripheral Blood Leukocytes Correlates with Intellectual Disability. Diagnostics (Basel) 2021; 11:diagnostics11101780. [PMID: 34679478 PMCID: PMC8534530 DOI: 10.3390/diagnostics11101780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/17/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. FXS is an X-linked, neurodevelopmental disorder caused by a CGG trinucleotide repeat expansion in the 5′ untranslated region (UTR) of the Fragile X Mental Retardation gene, FMR1. Greater than 200 CGG repeats results in epigenetic silencing of the gene leading to the deficiency or absence of Fragile X mental retardation protein (FMRP). The loss of FMRP is considered the root cause of FXS. The relationship between neurological function and FMRP expression in peripheral blood mononuclear cells (PBMCs) has not been well established. Assays to detect and measure FMR1 and FMRP have been described; however, none are sufficiently sensitive, precise, or quantitative to properly characterize the relationships between cognitive ability and CGG repeat number, FMR1 mRNA expression, or FMRP expression measured in PBMCs. To address these limitations, two novel immunoassays were developed and optimized, an electro-chemiluminescence immunoassay and a multiparameter flow cytometry assay. Both assays were performed on PMBCs isolated from 27 study participants with FMR1 CGG repeats ranging from normal to full mutation. After correcting for methylation, a significant positive correlation between CGG repeat number and FMR1 mRNA expression levels and a significant negative correlation between FMRP levels and CGG repeat expansion was observed. Importantly, a high positive correlation was observed between intellectual quotient (IQ) and FMRP expression measured in PBMCs.
Collapse
|
48
|
Grosso V, Marcolungo L, Maestri S, Alfano M, Lavezzari D, Iadarola B, Salviati A, Mariotti B, Botta A, D’Apice MR, Novelli G, Delledonne M, Rossato M. Characterization of FMR1 Repeat Expansion and Intragenic Variants by Indirect Sequence Capture. Front Genet 2021; 12:743230. [PMID: 34646309 PMCID: PMC8504923 DOI: 10.3389/fgene.2021.743230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022] Open
Abstract
Traditional methods for the analysis of repeat expansions, which underlie genetic disorders, such as fragile X syndrome (FXS), lack single-nucleotide resolution in repeat analysis and the ability to characterize causative variants outside the repeat array. These drawbacks can be overcome by long-read and short-read sequencing, respectively. However, the routine application of next-generation sequencing in the clinic requires target enrichment, and none of the available methods allows parallel analysis of long-DNA fragments using both sequencing technologies. In this study, we investigated the use of indirect sequence capture (Xdrop technology) coupled to Nanopore and Illumina sequencing to characterize FMR1, the gene responsible of FXS. We achieved the efficient enrichment (> 200×) of large target DNA fragments (~60-80 kbp) encompassing the entire FMR1 gene. The analysis of Xdrop-enriched samples by Nanopore long-read sequencing allowed the complete characterization of repeat lengths in samples with normal, pre-mutation, and full mutation status (> 1 kbp), and correctly identified repeat interruptions relevant for disease prognosis and transmission. Single-nucleotide variants (SNVs) and small insertions/deletions (indels) could be detected in the same samples by Illumina short-read sequencing, completing the mutational testing through the identification of pathogenic variants within the FMR1 gene, when no typical CGG repeat expansion is detected. The study successfully demonstrated the parallel analysis of repeat expansions and SNVs/indels in the FMR1 gene at single-nucleotide resolution by combining Xdrop enrichment with two next-generation sequencing approaches. With the appropriate optimization necessary for the clinical settings, the system could facilitate both the study of genotype-phenotype correlation in FXS and enable a more efficient diagnosis and genetic counseling for patients and their relatives.
Collapse
Affiliation(s)
- Valentina Grosso
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Luca Marcolungo
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Simone Maestri
- Department of Biotechnology, University of Verona, Verona, Italy
| | | | - Denise Lavezzari
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Barbara Iadarola
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Alessandro Salviati
- Department of Biotechnology, University of Verona, Verona, Italy
- GENARTIS srl, Verona, Italy
| | - Barbara Mariotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Annalisa Botta
- Department of Biomedicine and Prevention, Medical Genetics Section, University of Rome "Tor Vergata", Rome, Italy
| | | | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Medical Genetics Section, University of Rome "Tor Vergata", Rome, Italy
- IRCCS Neuromed Mediterranean Neurological Institute, Pozzilli, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Massimo Delledonne
- Department of Biotechnology, University of Verona, Verona, Italy
- GENARTIS srl, Verona, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
- GENARTIS srl, Verona, Italy
| |
Collapse
|
49
|
Salcedo-Arellano MJ, Sanchez D, Wang JY, McLennan YA, Clark CJ, Juarez P, Schneider A, Tassone F, Hagerman RJ, Martínez-Cerdeño V. Case Report: Coexistence of Alzheimer-Type Neuropathology in Fragile X-Associated Tremor Ataxia Syndrome. Front Neurosci 2021; 15:720253. [PMID: 34602969 PMCID: PMC8485779 DOI: 10.3389/fnins.2021.720253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Abstract
This case documents the co-occurrence of the fragile X-associated tremor ataxia syndrome (FXTAS) and Alzheimer-type neuropathology in a 71-year-old premutation carrier with 85 CGG repeats in the fragile X mental retardation 1 (FMR1) gene, in addition to an apolipoprotein E (APOE) ε4 allele. FXTAS and Alzheimer's Disease (AD) are late-onset neurodegenerative diseases that share overlapping cognitive deficits including processing speed, working memory and executive function. The prevalence of coexistent FXTAS-AD pathology remains unknown. The clinical picture in this case was marked with rapid cognitive decline between age 67 and 71 years in addition to remarkable MRI changes. Over the 16 months between the two clinical evaluations, the brain atrophied 4.12% while the lateral ventricles increased 26.4% and white matter hyperintensities (WMH) volume increased 15.6%. Other regions atrophied substantially faster than the whole brain included the thalamus (-6.28%), globus pallidus (-10.95%), hippocampus (-6.95%), and amygdala (-7.58%). A detailed postmortem assessment included an MRI with confluent WMH and evidence of cerebral microbleeds (CMB). The histopathological study demonstrated FXTAS inclusions in neurons and astrocytes, a widespread presence of phosphorylated tau protein and, amyloid β plaques in cortical areas and the hippocampus. CMBs were noticed in the precentral gyrus, middle temporal gyrus, visual cortex, and brainstem. There were high amounts of iron deposits in the globus pallidus and the putamen consistent with MRI findings. We hypothesize that coexistent FXTAS-AD neuropathology contributed to the steep decline in cognitive abilities.
Collapse
Affiliation(s)
- Maria Jimena Salcedo-Arellano
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, United States
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Desiree Sanchez
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Jun Yi Wang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
- Center for Mind and Brain, University of California, Davis, Davis, CA, United States
| | - Yingratana A. McLennan
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
| | - Courtney Jessica Clark
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
| | - Pablo Juarez
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Andrea Schneider
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, United States
| | - Randi J. Hagerman
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
| | - Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, United States
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| |
Collapse
|
50
|
Napoli E, Flores A, Mansuri Y, Hagerman RJ, Giulivi C. Sulforaphane improves mitochondrial metabolism in fibroblasts from patients with fragile X-associated tremor and ataxia syndrome. Neurobiol Dis 2021; 157:105427. [PMID: 34153466 PMCID: PMC8475276 DOI: 10.1016/j.nbd.2021.105427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 02/09/2023] Open
Abstract
CGG expansions between 55 and 200 in the 5'-untranslated region of the fragile-X mental retardation gene (FMR1) increase the risk of developing the late-onset debilitating neuromuscular disease Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). While the science behind this mutation, as a paradigm for RNA-mediated nucleotide triplet repeat expansion diseases, has progressed rapidly, no treatment has proven effective at delaying the onset or decreasing morbidity, especially at later stages of the disease. Here, we demonstrated the beneficial effect of the phytochemical sulforaphane (SFN), exerted through NRF2-dependent and independent manner, on pathways relevant to brain function, bioenergetics, unfolded protein response, proteosome, antioxidant defenses, and iron metabolism in fibroblasts from FXTAS-affected subjects at all disease stages. This study paves the way for future clinical studies with SFN in the treatment of FXTAS, substantiated by the established use of this agent in clinical trials of diseases with NRF2 dysregulation and in which age is the leading risk factor.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Amanda Flores
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616;,Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Yasmeen Mansuri
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Randi J. Hagerman
- Department of Pediatrics, University of California Davis Medical Center, Sacramento, CA;,Medical Investigations of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California Davis, CA 95817
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, United States of America; Medical Investigations of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California Davis, CA 95817, USA.
| |
Collapse
|