1
|
Vilkaite A, Nguyen XP, Güzel CT, Gottschlich L, Bender U, Dietrich JE, Hinderhofer K, Strowitzki T, Rehnitz J. Beyond Repetition: The Role of Gray Zone Alleles in the Upregulation of FMR1-Binding miR-323a-3p and the Modification of BMP/SMAD-Pathway Gene Expression in Human Granulosa Cells. Int J Mol Sci 2025; 26:3192. [PMID: 40244008 PMCID: PMC11989689 DOI: 10.3390/ijms26073192] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The Fragile X mental retardation type 1 gene (FMR1) contains a CGG triplet cluster of varied length (30 repeats on average) located in its 5' UTR. In its premutated state (54-200 repeats), FMR1 contributes to the pathogenesis of premature ovarian insufficiency (POI). Its gray zone alleles (41-54 repeats) are supposed to impair the ovarian function as well. In the case of a CGG repeat length > 200, Fragile X syndrome occurs. Post-transcriptional expression of FMR1 is regulated by microRNAs. Although miR-323a-3p overexpression suppresses FMR1 in various tissues, this relationship has not been evaluated in the human ovary. Additionally, this microRNA targets SMADs, which are suggested regulators of ovarian cell proliferation, growth, and function. This study investigated how FMR1 allele lengths with CGG repeat numbers n < 55 (normal and gray zone genotypes) relate to miR-323a-3p expression and how they may impact associated SMAD expression in human granulosa cells. COV434 cells and patient-derived GCs were used to evaluate FMR1, miR-323a-3p, and BMP/SMAD-pathway member expression levels. Briefly, miR-323a-3p was significantly upregulated in GCs of the gray zone group compared to the normal allele group (p < 0.0001), while the FMR1 level did not vary. Furthermore, the gray zone group showed a significant upregulation of BMPR2, SMAD1, SMAD4, and SMAD9. In contrast, the miR-323a-3p transfection of COV434 cells significantly downregulated SMAD3, SMAD4, SMAD5, and SMAD9, while the FMR1 and SMAD1 levels remained stable. Our findings highlight a CGG repeat number-dependent upregulation of miR-323a-3p and an alteration of the BMP/SMAD pathway, suggesting that these changes happen and contribute to impaired ovarian function independently.
Collapse
Affiliation(s)
- Adriana Vilkaite
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Xuan Phuoc Nguyen
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Cansu Türkan Güzel
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Lucas Gottschlich
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Ulrike Bender
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Jens E. Dietrich
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Katrin Hinderhofer
- Institute of Human Genetics, University Heidelberg, 69120 Heidelberg, Germany;
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Julia Rehnitz
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| |
Collapse
|
2
|
Mattingly Z, Chetty S. Untangling the Molecular Mechanisms Contributing to Autism Spectrum Disorder Using Stem Cells. Autism Res 2025; 18:476-485. [PMID: 39989339 DOI: 10.1002/aur.70005] [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: 08/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neuro developmental condition characterized by significant genetic and phenotypic variability, making diagnosis and treatment challenging. The heterogeneity of ASD-associated genetic variants and the absence of clear causal factors in many cases complicate personalized care. Traditional models, such as postmortem brain tissue and animal studies, have provided valuable insights but are limited in capturing the dynamic processes and human-specific aspects of ASD pathology. Recent advances in human induced pluripotent stem cell (iPSC) technology have transformed ASD research by enabling the generation of patient-derived neural cells in both two-dimensional cultures and three-dimensional brain organoid models. These models retain the donor's genetic background, allowing researchers to investigate disease-specific cellular and molecular mechanisms while identifying potential therapeutic targets tailored to individual patients. This commentary highlights how stem cell-based approaches are advancing our understanding of ASD and paving the way for more personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zoe Mattingly
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sundari Chetty
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Shirvani P, Shirvani A, Holick MF. Mitochondrial Dysfunction and Its Potential Molecular Interplay in Hypermobile Ehlers-Danlos Syndrome: A Scoping Review Bridging Cellular Energetics and Genetic Pathways. Curr Issues Mol Biol 2025; 47:134. [PMID: 39996855 PMCID: PMC11854588 DOI: 10.3390/cimb47020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Hypermobile Ehlers-Danlos Syndrome (hEDS) is a hereditary connective tissue disorder characterized by joint hypermobility, skin hyperextensibility, and systemic manifestations such as chronic fatigue, gastrointestinal dysfunction, and neurological symptoms. Unlike other EDS subtypes with known genetic mutations, hEDS lacks definitive markers, suggesting a multifactorial etiology involving both mitochondrial dysfunction and non-mitochondrial pathways. This scoping review, conducted in accordance with the PRISMA-ScR guidelines, highlights mitochondrial dysfunction as a potential unifying mechanism in hEDS pathophysiology. Impaired oxidative phosphorylation (OXPHOS), elevated reactive oxygen species (ROS) levels, and calcium dysregulation disrupt cellular energetics and extracellular matrix (ECM) homeostasis, contributing to the hallmark features of hEDS. We reviewed candidate genes associated with ECM remodeling, signaling pathways, and immune regulation. Protein-protein interaction (PPI) network analyses revealed interconnected pathways linking mitochondrial dysfunction with these candidate genes. Comparative insights from Fabry disease and fragile X premutation carriers underscore shared mechanisms such as RNA toxicity, matrix metalloproteinases (MMP) activation, and ECM degradation. These findings may suggest that mitochondrial dysfunction amplifies systemic manifestations through its interplay with non-mitochondrial molecular pathways. By integrating these perspectives, this review provides a potential framework for understanding hEDS pathogenesis while highlighting latent avenues for future research into its molecular basis. Understanding the potential role of mitochondrial dysfunction in hEDS not only sheds light on its complex molecular etiology but also opens new paths for targeted interventions.
Collapse
Affiliation(s)
| | - Arash Shirvani
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Michael F. Holick
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
4
|
García‐de Soto J, Pouso‐Diz JM, Fernández‐Pajarín G, Román‐Pena P, Álvarez‐Noval A, Blanco‐Ulla M, Arán‐Echabe E, Ares B, Sesar Á. MR-Guided Focused Ultrasound for the Treatment of Tremor in Fragile X-Associated Tremor/Ataxia Syndrome. Mov Disord Clin Pract 2025; 12:246-248. [PMID: 39569492 PMCID: PMC11802649 DOI: 10.1002/mdc3.14270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 10/03/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024] Open
Affiliation(s)
- Jesús García‐de Soto
- Department of NeurologyComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
| | | | - Gustavo Fernández‐Pajarín
- Department of NeurologyComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
- Grupo Clínico de Trastornos del Movimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)Santiago de CompostelaSpain
- Universidad de Santiago de Compostela (USC)Santiago de CompostelaSpain
| | - Paula Román‐Pena
- Department of NeurosurgeryComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
| | | | - Miguel Blanco‐Ulla
- Department of RadiologyComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
| | - Eduardo Arán‐Echabe
- Universidad de Santiago de Compostela (USC)Santiago de CompostelaSpain
- Department of NeurosurgeryComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
| | - Begoña Ares
- Department of NeurologyComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
- Grupo Clínico de Trastornos del Movimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)Santiago de CompostelaSpain
| | - Ángel Sesar
- Department of NeurologyComplejo Hospitalario UniversitarioSantiago de CompostelaSpain
- Grupo Clínico de Trastornos del Movimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)Santiago de CompostelaSpain
| |
Collapse
|
5
|
Gunapala KM, Gadban A, Noreen F, Schär P, Benvenisty N, Taylor V. Ascorbic Acid Ameliorates Molecular and Developmental Defects in Human-Induced Pluripotent Stem Cell and Cerebral Organoid Models of Fragile X Syndrome. Int J Mol Sci 2024; 25:12718. [PMID: 39684429 DOI: 10.3390/ijms252312718] [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: 10/03/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Fragile X Syndrome (FX) is the most common form of inherited cognitive impairment and falls under the broader category of Autism Spectrum Disorders (ASD). FX is caused by a CGG trinucleotide repeat expansion in the non-coding region of the X-linked Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene, leading to its hypermethylation and epigenetic silencing. Animal models of FX rely on the deletion of the Fmr1 gene, which fails to replicate the epigenetic silencing mechanism of the FMR1 gene observed in human patients. Human stem cells carrying FX repeat expansions have provided a better understanding of the basis of epigenetic silencing of FMR1. Previous studies have found that 5-Azacytidine (5Azac) can reverse this methylation; however, 5Azac can be toxic, which may limit its therapeutic potential. Here, we show that the dietary factor Ascorbic Acid (AsA) can reduce DNA methylation in the FMR1 locus and lead to an increase in FMR1 gene expression in FX iPSCs and cerebral organoids. In addition, AsA treatment rescued neuronal gene expression and morphological defects observed in FX iPSC-derived cerebral organoids. Hence, we demonstrate that the dietary co-factor AsA can partially revert the molecular and morphological defects seen in human FX models in vitro. Our findings have implications for the development of novel therapies for FX in the future.
Collapse
Affiliation(s)
- Keith M Gunapala
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Aseel Gadban
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Faiza Noreen
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 4031 Basel, Switzerland
| | - Primo Schär
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
6
|
Casula M, Marchetti D, Trevisan L, Pezzoli L, Bellini M, Patrone S, Zingarelli A, Gotta F, Iascone M, Mandich P. Genetics architecture of spontaneous coronary artery dissection in an Italian cohort. Front Cardiovasc Med 2024; 11:1486273. [PMID: 39654947 PMCID: PMC11625805 DOI: 10.3389/fcvm.2024.1486273] [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: 08/25/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024] Open
Abstract
Spontaneous coronary artery dissection (SCAD) is a relevant non-atherosclerotic cause of acute coronary syndrome with a complex genetic architecture. Recent discoveries have highlighted the potential role of miRNAs and protein-coding genes involved in the processing of small RNAs in the pathogenesis of SCAD. Furthermore, there may be a connection between SCAD and the increased cardiovascular risk observed in fragile X premutation carriers as well as a correlation with pathogenetic variants in genes encoding for collagen and extracellular matrix, which are related to connective tissue disorders (CTDs). In our cohort of 15 Italian SCAD patients, a total of 37 rare variants were identified in 34 genes using whole exome sequencing (WES) and TRIO-WES analysis when both parents were available. Three likely pathogenic/pathogenetic variants were found in genes previously associated with SCAD and CTDs (COL3A1, COL1A2, and SMAD3) and 26 variants of uncertain significance in genes previously associated with SCAD and CTDs. TRIO-WES analysis revealed 7 de novo variants, 1 of which was found in a potential novel candidate gene (DROSHA). In addition, a premutation allele of 55 ± 2 CGG repeats in the promoter of the FMR1 gene was identified in two related SCAD patients by test for CGG-repeat expansions in the 5'-UTR of the FMR1 gene. Our findings suggest various potential mechanisms such as mRNA toxicity, miRNA regulation, alteration of collagen, and the extracellular matrix architecture, all of which could disrupt vascular homeostasis, and finally, WES and TRIO-WES have proven to be the most powerful approaches for characterizing the genetic background of SCAD.
Collapse
Affiliation(s)
- Marta Casula
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Daniela Marchetti
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Lucia Trevisan
- Medical Genetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Pezzoli
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Matteo Bellini
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Serena Patrone
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Antonio Zingarelli
- Cardiological Unit, Ospedale Policlinico IRCSS San Martino, Genoa, Italy
| | - Fabio Gotta
- Medical Genetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Iascone
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paola Mandich
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Medical Genetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
7
|
Ramírez-Cheyne J, López D, Payán-Gómez C, Arcos-Burgos M, Saldarriaga W. Fragile X syndrome in the largest world clustering. I. Genetic epidemiology and founder effect outline. Am J Med Genet A 2024; 194:e63523. [PMID: 38164622 DOI: 10.1002/ajmg.a.63523] [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: 04/04/2023] [Revised: 09/19/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
The FMR1 5' regulation gene region harbors a CGG trinucleotide repeat expansion (CGG-TRE) that causes Fragile X syndrome (FXS) when it expands to more than 200 repetitions. Ricaurte is a small village in southwestern Colombia, with an FXS prevalence of 1 in 38 men and 1 in 100 women (~100 times higher than the worldwide reported prevalence), defining Ricaurte as the largest FXS cluster in the world. In the present study, using next-generation sequencing of whole exome capture, we genotype 55 individuals from Ricaurte (49 with either full mutation or with premutation), four individuals from neighboring villages (with either the full mutation or with the premutation), and one unaffected woman, native of Ricaurte, who did not belong to any of the affected families. With advanced clustering and haplotype reconstruction, we modeled a common haplotype of 33 SNPs spanning 83,567,899 bp and harboring the FMR1 gene. This reconstructed haplotype was found in all the men from Ricaurte who carried the expansion, demonstrating that the genetic conglomerate of FXS in this population is due to a founder effect. The definition of this founder effect and its population outlining will allow a better prediction, follow-up, precise and personalized characterization of epidemiological parameters, better knowledge of the disease's natural history, and confident improvement of the clinical attention, life quality, and health interventions for this community.
Collapse
Affiliation(s)
| | - Diana López
- Department of Biological Science, Faculty of Agricultural Sciences, Universidad Nacional de Colombia, Palmira, Colombia
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia. Sede de La Paz. Cesar, Colombia
| | - Mauricio Arcos-Burgos
- Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | | |
Collapse
|
8
|
Luermans J, Fleming J, O'Shea R, Barlow-Stewart K, Palmer EE, Leffler M. "We are not a typical family anymore": Exploring the experiences and support needs of fathers of children with Fragile X syndrome in Australia. Am J Med Genet A 2024; 194:e63470. [PMID: 37974553 DOI: 10.1002/ajmg.a.63470] [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: 06/21/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
A diagnosis of the X-linked condition Fragile X syndrome (FXS) in a child commonly reveals the mother's carrier status. Previous research focused on the genetic counseling process for the child and maternal family, despite calls for more research on the support needs of fathers. This study explored experiences and support needs of fathers at least 1 year after their child's FXS diagnosis to understand barriers and enablers and optimize health outcomes for the family. In-depth interviews were conducted with 11 fathers recruited through the Australian Genetics of Learning Disability Service and the Fragile X Association. Deidentified transcripts were analyzed using thematic analysis guided by an inductive approach. Four themes emerged: (1) making life easier through understanding-yesterday and today, (2) the path to a new normal-today and tomorrow, (3) seeking information and support, and (4) what men want. Fathers reported diagnostic odysseys, postdiagnostic grief, and challenges adjusting. They highlighted difficulties in understanding their child's unique behaviors and needs, responding to their partner's psychological support needs, planning for their child's future, and navigating complex health and disability systems. Participants suggested health professionals facilitate father-to-father support and psychological counseling. These findings highlight the unmet needs of fathers and suggest that a strengths-based approach is critically important given the recognized mental health impact.
Collapse
Affiliation(s)
- Jacintha Luermans
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia
| | - Jane Fleming
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia
| | - Rosie O'Shea
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia
| | - Kristine Barlow-Stewart
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, New South Wales, Australia
| | - Elizabeth Emma Palmer
- Discipline of Paediatrics and Child Health, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Melanie Leffler
- Genetics of Learning Disability (GOLD) Service, Hunter Genetics, Waratah, New South Wales, Australia
| |
Collapse
|
9
|
Hessl D, Rojas KM, Ferrer E, Espinal G, Famula J, Schneider A, Hagerman R, Tassone F, Rivera SM. FMR1 Carriers Report Executive Function Changes Prior to Fragile X-Associated Tremor/Ataxia Syndrome: A Longitudinal Study. Mov Disord 2024; 39:519-525. [PMID: 38124331 PMCID: PMC11268876 DOI: 10.1002/mds.29695] [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: 09/08/2023] [Revised: 11/06/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Men with fragile X-associated tremor/ataxia syndrome (FXTAS) often develop executive dysfunction, characterized by disinhibition, frontal dyscontrol of movement, and working memory and attention changes. Although cross-sectional studies have suggested that earlier executive function changes may precede FXTAS, the lack of longitudinal studies has made it difficult to address this hypothesis. OBJECTIVE To determine whether executive function deterioration experienced by premutation carriers (PC) in daily life precedes and predicts FXTAS. METHODS This study included 66 FMR1 PC ranging from 40 to 78 years (mean, 59.5) and 31 well-matched healthy controls (HC) ages 40 to 75 (mean, 57.7) at baseline. Eighty-four participants returned for 2 to 5 follow up visits over a duration of 1 to 9 years (mean, 4.6); 28 of the PC developed FXTAS. The Behavior Rating Inventory of Executive Function-Adult Version (BRIEF-A) was completed by participants and their spouses/partners at each visit. RESULTS Longitudinal mixed model regression analyses showed a greater decline with age in PC compared to HC on the Metacognition Index (MI; self-initiation, working memory, organization, task monitoring). Conversion to FXTAS was associated with worsening MI and Behavioral Regulation Index (BRI; inhibition, flexibility, emotion modulation). For spouse/partner report, FXTAS conversion was associated with worsening MI. Finally, increased self-report executive function problems at baseline significantly predicted later development of FXTAS. CONCLUSIONS Executive function changes experienced by male PC represent a prodrome of the later movement disorder. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- David Hessl
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Karina Mandujano Rojas
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Center for Mind and Brain, University of California Davis, Davis, California, USA
| | - Emilio Ferrer
- Department of Psychology, University of California Davis, Davis, CA, USA
| | - Glenda Espinal
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jessica Famula
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
- Family Caregiving Institute, Betty Irene Moore School of Nursing, University of California Davis, Sacramento, California, USA
| | - Andrea Schneider
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Flora Tassone
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Davis, California, USA
| | - Susan M. Rivera
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychology, University of California Davis, Davis, CA, USA
- Center for Mind and Brain, University of California Davis, Davis, California, USA
- Department of Psychology, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
10
|
Persico T, Tranquillo ML, Seracchioli R, Zuccarello D, Sorrentino U. PGT-M for Premature Ovarian Failure Related to CGG Repeat Expansion of the FMR1 Gene. Genes (Basel) 2023; 15:6. [PMID: 38275588 PMCID: PMC10815814 DOI: 10.3390/genes15010006] [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: 10/25/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Primary ovarian failure (POF) is caused by follicle exhaustion and is associated with menstrual irregularities and elevated gonadotropin levels, which lead to infertility before the age of 40 years. The etiology of POI is mostly unknown, but a heterogeneous genetic and familial background can be identified in a subset of cases. Abnormalities in the fragile X mental retardation 1 gene (FMR1) are among the most prevalent monogenic causes of POI. These abnormalities are caused by the expansion of an unstable CGG repeat in the 5' untranslated region of FMR1. Expansions over 200 repeats cause fragile X syndrome (FXS), whereas expansions between 55 and 200 CGG repeats, which are defined as a fragile X premutation, have been associated with premature ovarian failure type 1 (POF1) in heterozygous females. Preimplantation genetic testing for monogenic diseases (PGT-M) can be proposed when the female carries a premutation or a full mutation. In this narrative review, we aim to recapitulate the clinical and molecular features of POF1 and their implications in the context of PGT-M.
Collapse
Affiliation(s)
- Tiziana Persico
- Medically Assisted Procreation Center, Maternal and Child Department, Beauregard Hospital, Valle D’Aosta Local Public Health, 11100 Aoste, Italy
| | - Maria Lucrezia Tranquillo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (M.L.T.); (R.S.)
| | - Renato Seracchioli
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (M.L.T.); (R.S.)
- Division of Gynaecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero, University of Bologna, 40138 Bologna, Italy
| | - Daniela Zuccarello
- Clinical Genetics and Epidemiology Unit, University of Padova, 35128 Padova, Italy; (D.Z.); (U.S.)
| | - Ugo Sorrentino
- Clinical Genetics and Epidemiology Unit, University of Padova, 35128 Padova, Italy; (D.Z.); (U.S.)
| |
Collapse
|
11
|
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
|
12
|
Alvarez-Mora MI, Garrabou G, Molina-Porcel L, Grillo-Risco R, Garcia-Garcia F, Barcos T, Cantó-Santos J, Rodriguez-Revenga L. Exploration of SUMO2/3 Expression Levels and Autophagy Process in Fragile X-Associated Tremor/Ataxia Syndrome: Addressing Study Limitations and Insights for Future Research. Cells 2023; 12:2364. [PMID: 37830578 PMCID: PMC10571773 DOI: 10.3390/cells12192364] [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: 07/20/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder that appears in adult FMR1 premutation carriers. The neuropathological hallmark of FXTAS is an intranuclear inclusion in neurons and astrocytes. Nearly 200 different proteins have been identified in FXTAS inclusions, being the small ubiquitin-related modifier 2 (SUMO2), ubiquitin and p62 the most highly abundant. These proteins are components of the protein degradation machinery. This study aimed to characterize SUMO2/3 expression levels and autophagy process in human postmortem brain samples and skin fibroblast cultures from FXTAS patients. Results revealed that FXTAS postmortem brain samples are positive for SUMO2/3 conjugates and supported the idea that SUMO2/3 accumulation is involved in inclusion formation. Insights from RNA-sequencing data indicated that SUMOylation processes are significantly upregulated in FXTAS samples. In addition, the analysis of the autophagy flux showed the accumulation of p62 protein levels and autophagosomes in skin fibroblasts from FXTAS patients. Similarly, gene set analysis evidenced a significant downregulation in gene ontology terms related to autophagy in FXTAS samples. Overall, this study provides new evidence supporting the role of SUMOylation and autophagic processes in the pathogenic mechanisms underlying FXTAS.
Collapse
Affiliation(s)
- Maria Isabel Alvarez-Mora
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.I.A.-M.); (T.B.)
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, 08036 Barcelona, Spain; (G.G.)
- Fundacio de Recerca Clínic Barcelona-Institut d’Investigacions Biomediques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain;
| | - Glòria Garrabou
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, 08036 Barcelona, Spain; (G.G.)
- Inherited Metabolic Diseases and Muscle Disorders’ Research Laboratory (U722), Cellex-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, Internal Medicine Department––Hospital Clínic Clinic of Barcelona, 08036 Barcelona, Spain
| | - Laura Molina-Porcel
- Fundacio de Recerca Clínic Barcelona-Institut d’Investigacions Biomediques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain;
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clinic, 08036 Barcelona, Spain
- Neurological Tissue Bank of the Biobanc-Hospital Clinic-FCRB-IDIBAPS, 08036 Barcelona, Spain
| | - Ruben Grillo-Risco
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain; (R.G.-R.); (F.G.-G.)
| | - Francisco Garcia-Garcia
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain; (R.G.-R.); (F.G.-G.)
| | - Tamara Barcos
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.I.A.-M.); (T.B.)
| | - Judith Cantó-Santos
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, 08036 Barcelona, Spain; (G.G.)
- Inherited Metabolic Diseases and Muscle Disorders’ Research Laboratory (U722), Cellex-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, Internal Medicine Department––Hospital Clínic Clinic of Barcelona, 08036 Barcelona, Spain
| | - Laia Rodriguez-Revenga
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.I.A.-M.); (T.B.)
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, 08036 Barcelona, Spain; (G.G.)
- Fundacio de Recerca Clínic Barcelona-Institut d’Investigacions Biomediques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain;
| |
Collapse
|
13
|
Hessl D, Rojas KM, Ferrer E, Espinal G, Famula J, Schneider A, Elagerman R, Tassone F, Rivera SM. A Longitudinal Study of Executive Function in Daily Life in Male Fragile X Premutation Carriers and Association with FXTAS Conversion. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.31.23294855. [PMID: 37693384 PMCID: PMC10491369 DOI: 10.1101/2023.08.31.23294855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Background Men with fragile X-associated tremor/ataxia syndrome (FXTAS) often develop executive dysfunction, characterized by disinhibition, frontal dyscontrol of movement, and working memory and attention changes. Although cross-sectional studies have suggested that earlier executive function changes may precede FXTAS, the lack of longitudinal studies have made it difficult to address this hypothesis. Methods This study included 66 FMR1 premutation carriers (PC) ranging from 40-78 years (Mean=59.5) and 31 well-matched healthy controls (HC) ages 40-75 (Mean 57.7) at baseline. Eighty-four participants returned for 2-5 follow up visits over a duration of 1 to 9 years (Mean=4.6); 28 of the PC developed FXTAS. The Behavior Rating Inventory of Executive Function-Adult Version (BRIEF-A) was completed by participants and their spouses/partners at each visit. Results Longitudinal mixed model regression analyses showed a greater decline with age in PC compared to HC on the Metacognition Index (MI; self-initiation, working memory, organization, task monitoring). Conversion to FXTAS was associated with worsening MI and Behavioral Regulation Index (BRI; inhibition, flexibility, emotion modulation). For spouse/partner report, FXTAS conversion was associated with worsening MI. Finally, BRIEF-A executive function problems at baseline significantly predicted later development of FXTAS. Conclusions These findings suggest that executive function changes represent a prodrome of the later movement disorder.
Collapse
Affiliation(s)
- David Hessl
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Karina Mandujano Rojas
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Center for Mind and Brain, University of California Davis, Davis, California, USA
| | - Emilio Ferrer
- Department of Psychology, University of California Davis, Davis, CA, USA
| | - Glenda Espinal
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jessica Famula
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
- Family Caregiving Institute, Betty Irene Moore School of Nursing, University of California Davis, Sacramento, California, USA
| | - Andrea Schneider
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Randi Elagerman
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Flora Tassone
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Davis, California, USA
| | - Susan M. Rivera
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Psychology, University of California Davis, Davis, CA, USA
- Center for Mind and Brain, University of California Davis, Davis, California, USA
- Department of Psychology, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
14
|
Friedman L, Lauber M, Behroozmand R, Fogerty D, Kunecki D, Berry-Kravis E, Klusek J. Atypical vocal quality in women with the FMR1 premutation: an indicator of impaired sensorimotor control. Exp Brain Res 2023; 241:1975-1987. [PMID: 37347418 PMCID: PMC10863608 DOI: 10.1007/s00221-023-06653-2] [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: 02/07/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023]
Abstract
Women with the FMR1 premutation are susceptible to motor involvement related to atypical cerebellar function, including risk for developing fragile X tremor ataxia syndrome. Vocal quality analyses are sensitive to subtle differences in motor skills but have not yet been applied to the FMR1 premutation. This study examined whether women with the FMR1 premutation demonstrate differences in vocal quality, and whether such differences relate to FMR1 genetic, executive, motor, or health features of the FMR1 premutation. Participants included 35 women with the FMR1 premutation and 45 age-matched women without the FMR1 premutation who served as a comparison group. Three sustained /a/ vowels were analyzed for pitch (mean F0), variability of pitch (standard deviation of F0), and overall vocal quality (jitter, shimmer, and harmonics-to-noise ratio). Executive, motor, and health indices were obtained from direct and self-report measures and genetic samples were analyzed for FMR1 CGG repeat length and activation ratio. Women with the FMR1 premutation had a lower pitch, larger pitch variability, and poorer vocal quality than the comparison group. Working memory was related to harmonics-to-noise ratio and shimmer in women with the FMR1 premutation. Vocal quality abnormalities differentiated women with the FMR1 premutation from the comparison group and were evident even in the absence of other clinically evident motor deficits. This study supports vocal quality analyses as a tool that may prove useful in the detection of early signs of motor involvement in this population.
Collapse
Affiliation(s)
- Laura Friedman
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, USA
| | - Meagan Lauber
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, USA
| | - Roozbeh Behroozmand
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, USA
| | - Daniel Fogerty
- Department of Speech and Hearing Science, University of Illinois Urbana-Champaign, Champaign, USA
| | - Dariusz Kunecki
- Department of Pediatrics, Rush University Medical Center, Chicago, USA
| | | | - Jessica Klusek
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, USA.
| |
Collapse
|
15
|
Benussi A, Batsikadze G, França C, Cury RG, Maas RPPWM. The Therapeutic Potential of Non-Invasive and Invasive Cerebellar Stimulation Techniques in Hereditary Ataxias. Cells 2023; 12:cells12081193. [PMID: 37190102 DOI: 10.3390/cells12081193] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
The degenerative ataxias comprise a heterogeneous group of inherited and acquired disorders that are characterized by a progressive cerebellar syndrome, frequently in combination with one or more extracerebellar signs. Specific disease-modifying interventions are currently not available for many of these rare conditions, which underscores the necessity of finding effective symptomatic therapies. During the past five to ten years, an increasing number of randomized controlled trials have been conducted examining the potential of different non-invasive brain stimulation techniques to induce symptomatic improvement. In addition, a few smaller studies have explored deep brain stimulation (DBS) of the dentate nucleus as an invasive means to directly modulate cerebellar output, thereby aiming to alleviate ataxia severity. In this paper, we comprehensively review the clinical and neurophysiological effects of transcranial direct current stimulation (tDCS), repetitive transcranial magnetic stimulation (rTMS), and dentate nucleus DBS in patients with hereditary ataxias, as well as the presumed underlying mechanisms at the cellular and network level and perspectives for future research.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Giorgi Batsikadze
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, University of Duisburg-Essen, 45147 Essen, Germany
| | - Carina França
- Movement Disorders Center, Department of Neurology, University of São Paulo, São Paulo 05508-010, Brazil
| | - Rubens G Cury
- Movement Disorders Center, Department of Neurology, University of São Paulo, São Paulo 05508-010, Brazil
| | - Roderick P P W M Maas
- Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
16
|
Ishiura H, Tsuji S, Toda T. Recent advances in CGG repeat diseases and a proposal of fragile X-associated tremor/ataxia syndrome, neuronal intranuclear inclusion disease, and oculophryngodistal myopathy (FNOP) spectrum disorder. J Hum Genet 2023; 68:169-174. [PMID: 36670296 PMCID: PMC9968658 DOI: 10.1038/s10038-022-01116-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 01/22/2023]
Abstract
While whole genome sequencing and long-read sequencing have become widely available, more and more focuses are on noncoding expanded repeats. Indeed, more than half of noncoding repeat expansions related to diseases have been identified in the five years. An exciting aspect of the progress in this field is an identification of a phenomenon called repeat motif-phenotype correlation. Repeat motif-phenotype correlation in noncoding repeat expansion diseases is first found in benign adult familial myoclonus epilepsy. The concept is extended in the research of CGG repeat expansion diseases. In this review, we focus on newly identified CGG repeat expansion diseases, update the concept of repeat motif-phenotype correlation in CGG repeat expansion diseases, and propose a clinical concept of FNOP (fragile X-associated tremor/ataxia syndrome, neuronal intranuclear inclusion disease, and oculopharyngodistal myopathy)-spectrum disorder, which shares clinical features and thus probably share some common disease pathophysiology, to further facilitate discussion and progress in this field.
Collapse
Affiliation(s)
- Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Institute of Medical Genomics, International University of Health and Welfare, Narita, Japan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
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
|
18
|
Hong J, Dembo RS, DaWalt LS, Brilliant M, Berry-Kravis EM, Mailick M. The effect of college degree attainment on neurodegenerative symptoms in genetically at-risk women. SSM Popul Health 2022; 19:101262. [PMID: 36238818 PMCID: PMC9550653 DOI: 10.1016/j.ssmph.2022.101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 12/02/2022] Open
Abstract
Using longitudinal data, the present study examined the association between college degree attainment and the manifestation of neurodegenerative symptoms among women (n = 93) at elevated genetic risk. The neurodegenerative symptoms investigated in this study are due to FXTAS (Fragile X-associated Tremor/Ataxia Syndrome), a condition with onset after age 50. Those at risk for FXTAS have a mutation of a single gene found on the X chromosome. FXTAS is characterized by intention tremor, gait ataxia, executive function deficits, memory issues, and neuropathy. College degree attainment has been shown to provide neuroprotective effects in the general population, delaying the development of neurodegenerative conditions such as Alzheimer's disease. For this reason, college degree attainment is a potentially salient resource for those at risk of FXTAS. The results of the present research indicated significantly more severe FXTAS symptoms in women who did not attain a college degree as compared with those who were college graduates, although the two groups were similar in age, genetic risk, household income, health behaviors, and general health problems. Furthermore, symptoms in those who did not attain a college degree worsened over the 9-year study period at a significantly faster rate than the college graduates. The association between college degree attainment and FXTAS symptoms was significantly mediated by depression, which was lower among the graduates than those who did not attain a college degree. Thus, the present research is an example of how a sociodemographic factor can mitigate neurodegenerative conditions in genetically at-risk adults.
Collapse
Affiliation(s)
- Jinkuk Hong
- Waisman Center, University of Wisconsin-Madison, USA
| | | | | | | | | | | |
Collapse
|
19
|
Standlee J, Malkani R. Sleep Dysfunction in Movement Disorders: a Window to the Disease Biology. Curr Neurol Neurosci Rep 2022; 22:565-576. [PMID: 35867306 DOI: 10.1007/s11910-022-01220-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW To comprehensively summarize the sleep pathologies associated with movement disorders, focusing on neurodegenerative diseases. RECENT FINDINGS Mounting evidence has further implicated both sleep and circadian disruption in the pathophysiology of many movement disorders. In particular, recent data illuminate the mechanisms by which poor sleep quality and circadian dysfunction can exacerbate neurodegeneration. In addition, anti-IgLON5 disease is a recently described autoimmune disease with various symptoms that can feature prominent sleep disruption and parasomnia. Many movement disorders are associated with sleep and circadian rhythm disruption. Motor symptoms can cause sleep fragmentation, resulting in insomnia and excessive daytime sleepiness. Many neurodegenerative movement disorders involve brainstem pathology in regions close to or affecting nuclei that regulate sleep and wake. Further, commonly used movement medications may exacerbate sleep concerns. Providers should screen for and address these sleep symptoms to improve function and quality of life for patients and caregivers.
Collapse
Affiliation(s)
- Jordan Standlee
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Roneil Malkani
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Jesse Brown Veterans Affairs Medical Center, Neurology Service, 820 S Damen Ave, Damen Building, 9th floor, Chicago, IL, 60612, USA.
| |
Collapse
|
20
|
Hwang JY, Monday HR, Yan J, Gompers A, Buxbaum AR, Sawicka KJ, Singer RH, Castillo PE, Zukin RS. CPEB3-dependent increase in GluA2 subunits impairs excitatory transmission onto inhibitory interneurons in a mouse model of fragile X. Cell Rep 2022; 39:110853. [PMID: 35675768 PMCID: PMC9671216 DOI: 10.1016/j.celrep.2022.110853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/05/2021] [Accepted: 05/01/2022] [Indexed: 01/29/2023] Open
Abstract
Fragile X syndrome (FXS) is a leading cause of inherited intellectual disability and autism. Whereas dysregulated RNA translation in Fmr1 knockout (KO) mice, a model of FXS, is well studied, little is known about aberrant transcription. Using single-molecule mRNA detection, we show that mRNA encoding the AMPAR subunit GluA2 (but not GluA1) is elevated in dendrites and at transcription sites of hippocampal neurons of Fmr1 KO mice, indicating elevated GluA2 transcription. We identify CPEB3, a protein implicated in memory consolidation, as an upstream effector critical to GluA2 mRNA expression in FXS. Increased GluA2 mRNA is translated into an increase in GluA2 subunits, a switch in synaptic AMPAR phenotype from GluA2-lacking, Ca2+-permeable to GluA2-containing, Ca2+-impermeable, reduced inhibitory synaptic transmission, and loss of NMDAR-independent LTP at glutamatergic synapses onto CA1 inhibitory interneurons. These factors could contribute to an excitatory/inhibitory imbalance-a common theme in FXS and other autism spectrum disorders.
Collapse
Affiliation(s)
- Jee-Yeon Hwang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA,These authors contributed equally,Lead contact,Correspondence: (J.-Y.H.), (R.S.Z.)
| | - Hannah R. Monday
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Present address: Department of Molecular and Cellular Biology and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA,These authors contributed equally
| | - Jingqi Yan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA,These authors contributed equally
| | - Andrea Gompers
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA 95616, USA,These authors contributed equally
| | - Adina R. Buxbaum
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Structural & Cell Biology, Albert Einstein College of Medicine, New York, NY 10461, USA,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA,Present address: Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kirsty J. Sawicka
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Present address: Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Robert H. Singer
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Structural & Cell Biology, Albert Einstein College of Medicine, New York, NY 10461, USA,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA,These authors contributed equally
| | - Pablo E. Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, NY 10461, USA,These authors contributed equally
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,These authors contributed equally,Correspondence: (J.-Y.H.), (R.S.Z.)
| |
Collapse
|
21
|
Boldyreva LV, Andreyeva EN, Pindyurin AV. Position Effect Variegation: Role of the Local Chromatin Context in Gene Expression Regulation. Mol Biol 2022. [DOI: 10.1134/s0026893322030049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Talman LS, Pfeiffer RF. Movement Disorders and the Gut: A Review. Mov Disord Clin Pract 2022; 9:418-428. [PMID: 35586541 PMCID: PMC9092751 DOI: 10.1002/mdc3.13407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/07/2022] Open
Abstract
There is a close link between multiple movement disorders and gastrointestinal dysfunction. Gastrointestinal symptoms may precede the development of the neurologic syndrome or may arise following the neurologic presentation. This review will provide an overview of gastrointestinal accompaniments to several well-known as well as lesser known movement disorders. It will also highlight several disorders which may not be considered primary movement disorders but have an overlapping presentation of both gastrointestinal and movement abnormalities.
Collapse
Affiliation(s)
- Lauren S. Talman
- Department of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - Ronald F. Pfeiffer
- Department of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
23
|
Zhang S, Shen L, Jiao B. Cognitive Dysfunction in Repeat Expansion Diseases: A Review. Front Aging Neurosci 2022; 14:841711. [PMID: 35478698 PMCID: PMC9036481 DOI: 10.3389/fnagi.2022.841711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
With the development of the sequencing technique, more than 40 repeat expansion diseases (REDs) have been identified during the past two decades. Moreover, the clinical features of these diseases show some commonality, and the nervous system, especially the cognitive function was affected in part by these diseases. However, the specific cognitive domains impaired in different diseases were inconsistent. Here, we survey literature on the cognitive consequences of the following disorders presenting cognitive dysfunction and summarizing the pathogenic genes, epidemiology, and different domains affected by these diseases. We found that the cognitive domains affected in neuronal intranuclear inclusion disease (NIID) were widespread including the executive function, memory, information processing speed, attention, visuospatial function, and language. Patients with C9ORF72-frontotemporal dementia (FTD) showed impairment in executive function, memory, language, and visuospatial function. While in Huntington's disease (HD), the executive function, memory, and information processing speed were affected, in the fragile X-associated tremor/ataxia syndrome (FXTAS), executive function, memory, information processing speed, and attention were impaired. Moreover, the spinocerebellar ataxias showed broad damage in almost all the cognitive domains except for the relatively intact language ability. Some other diseases with relatively rare clinical data also indicated cognitive dysfunction, such as myotonic dystrophy type 1 (DM1), progressive myoclonus epilepsy (PME), Friedreich ataxia (FRDA), Huntington disease like-2 (HDL2), and cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS). We drew a cognitive function landscape of the related REDs that might provide an aspect for differential diagnosis through cognitive domains and effective non-specific interventions for these diseases.
Collapse
Affiliation(s)
- Sizhe Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- *Correspondence: Bin Jiao
| |
Collapse
|
24
|
Orsucci D, Lorenzetti L, Baldinotti F, Rossi A, Vitolo E, Gheri FL, Napolitano A, Tintori G, Vista M. Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS): A Gender Perspective. J Clin Med 2022; 11:jcm11041002. [PMID: 35207276 PMCID: PMC8876035 DOI: 10.3390/jcm11041002] [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: 12/15/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 11/22/2022] Open
Abstract
Although larger trinucleotide expansions give rise to a neurodevelopmental disorder called fragile X syndrome, fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder caused by a “premutation” (55–200 CGG repeats) in the FMR1 gene. FXTAS is one of the more common single-gene forms of late-onset ataxia and tremor that may have a more complex development in women, with atypical presentations. After a brief presentation of the atypical case of an Italian woman with FXTAS, who had several paroxysmal episodes suggestive of acute cerebellar and/or brainstem dysfunction, this article will revise the phenotype of FXTAS in women. Especially in females, FXTAS has a broad spectrum of symptoms, ranging from relatively severe diseases in mid-adulthood to mild cases beginning in later life. Female FXTAS and male FXTAS have a different symptomatic spectrum, and studies on the fragile X premutation should be conducted separately on women or men. Hopefully, a better understanding of the molecular processes involved in the polymorphic features of FXTAS will lead to more specific and effective therapies for this complex disorder.
Collapse
Affiliation(s)
- Daniele Orsucci
- Unit of Neurology, San Luca Hospital, Via Lippi-Francesconi, 55100 Lucca, Italy;
- Correspondence: or
| | - Lucia Lorenzetti
- Unit of Internal Medicine, Santa Croce Hospital, 55032 Castelnuovo Garfagnana, Lucca, Italy; (L.L.); (E.V.); (F.L.G.); (G.T.)
| | - Fulvia Baldinotti
- Laboratory of Molecular Genetics, University Hospital of Pisa, 56126 Pisa, Italy;
| | - Andrea Rossi
- Medical Affairs and Scientific Communications, 1260 Nyon, Switzerland;
| | - Edoardo Vitolo
- Unit of Internal Medicine, Santa Croce Hospital, 55032 Castelnuovo Garfagnana, Lucca, Italy; (L.L.); (E.V.); (F.L.G.); (G.T.)
| | - Fabio Luigi Gheri
- Unit of Internal Medicine, Santa Croce Hospital, 55032 Castelnuovo Garfagnana, Lucca, Italy; (L.L.); (E.V.); (F.L.G.); (G.T.)
| | | | - Giancarlo Tintori
- Unit of Internal Medicine, Santa Croce Hospital, 55032 Castelnuovo Garfagnana, Lucca, Italy; (L.L.); (E.V.); (F.L.G.); (G.T.)
| | - Marco Vista
- Unit of Neurology, San Luca Hospital, Via Lippi-Francesconi, 55100 Lucca, Italy;
| |
Collapse
|
25
|
Behavior Problems and Social Competence in Fragile X Syndrome: A Systematic Review. Genes (Basel) 2022; 13:genes13020280. [PMID: 35205326 PMCID: PMC8871871 DOI: 10.3390/genes13020280] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) causes intellectual disability and is the known leading cause of autism. Common problems in FXS include behavior and social problems. Along with syndromic characteristics and autism comorbidity, environmental factors might influence these difficulties. This systematic review focuses on the last 20 years of studies concerning behavior and social problems in FXS, considering environmental and personal variables that might influence both problems. Three databases were reviewed, leading to fifty-one studies meeting the inclusion criteria. Attention deficit hyperactivity disorder (ADHD) problems remain the greatest behavior problems, with behavioral problems and social competence being stable during the 20 years. Some developmental trajectories might have changed due to higher methodological control, such as aggressive behavior and attention problems. The socialization trajectory from childhood to adolescence remains unclear. Comorbidity with autism in individuals with FXS increased behavior problems and worsened social competence profiles. At the same time, comparisons between individuals with comorbid FXS and autism and individuals with autism might help define the comorbid phenotype. Environmental factors and parental characteristics influenced behavior problems and social competence. Higher methodological control is needed in studies including autism symptomatology and parental characteristics. More studies comparing autism in FXS with idiopathic autism are needed to discern differences between conditions.
Collapse
|
26
|
Schmitt LM, Dominick KC, Liu R, Pedapati EV, Ethridge LE, Smith E, Sweeney JA, Erickson CA. Evidence for Three Subgroups of Female FMR1 Premutation Carriers Defined by Distinct Neuropsychiatric Features: A Pilot Study. Front Integr Neurosci 2022; 15:797546. [PMID: 35046780 PMCID: PMC8763356 DOI: 10.3389/fnint.2021.797546] [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: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 01/06/2023] Open
Abstract
Over 200 Cytosine-guanine-guanine (CGG) trinucleotide repeats in the 5' untranslated region of the Fragile X mental retardation 1 (FMR1) gene results in a "full mutation," clinically Fragile X Syndrome (FXS), whereas 55 - 200 repeats result in a "premutation." FMR1 premutation carriers (PMC) are at an increased risk for a range of psychiatric, neurocognitive, and physical conditions. Few studies have examined the variable expression of neuropsychiatric features in female PMCs, and whether heterogeneous presentation among female PMCs may reflect differential presentation of features in unique subgroups. In the current pilot study, we examined 41 female PMCs (ages 17-78 years) and 15 age-, sex-, and IQ-matched typically developing controls (TDC) across a battery of self-report, eye tracking, expressive language, neurocognitive, and resting state EEG measures to determine the feasibility of identifying discrete clusters. Secondly, we sought to identify the key features that distinguished these clusters of female PMCs. We found a three cluster solution using k-means clustering. Cluster 1 represented a psychiatric feature group (27% of our sample); cluster 2 represented a group with executive dysfunction and elevated high frequency neural oscillatory activity (32%); and cluster 3 represented a relatively unaffected group (41%). Our findings indicate the feasibility of using a data-driven approach to identify naturally occurring clusters in female PMCs using a multi-method assessment battery. CGG repeat count and its association with neuropsychiatric features differ across clusters. Together, our findings provide important insight into potential diverging pathophysiological mechanisms and risk factors for each female PMC cluster, which may ultimately help provide novel and individualized targets for treatment options.
Collapse
Affiliation(s)
- Lauren M. Schmitt
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Kelli C. Dominick
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Rui Liu
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Ernest V. Pedapati
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Lauren E. Ethridge
- Department of Psychology, University of Oklahoma, Norman, OK, United States
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Elizabeth Smith
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - John A. Sweeney
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Craig A. Erickson
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
27
|
Yorns WR. Neurologic Disorders Affecting the Foot and Ankle. Clin Podiatr Med Surg 2022; 39:15-35. [PMID: 34809793 DOI: 10.1016/j.cpm.2021.08.005] [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] [Indexed: 11/17/2022]
Abstract
The neurologic causes of foot and leg dysfunction are reviewed. Disorders causing foot and ankle pain, weakness, or other sensorimotor disturbances often cause difficulty with ambulation and prompt patients to seek medical evaluation. Physical signs and symptoms along with targeted diagnostic testing are needed to come to the correct diagnosis and treatment plan. An overview of peripheral nerve, muscle, and central nervous system disorders affecting the foot and leg are discussed.
Collapse
Affiliation(s)
- William R Yorns
- Department of Neurology, UCONN School of Medicine, Connecticut Children's Medical Center, 505 Farmington Avenue., 2nd Floor, Farmington, CT 06032, USA.
| |
Collapse
|
28
|
Keil Stietz KP, Sethi S, Klocke CR, de Ruyter TE, Wilson MD, Pessah IN, Lein PJ. Sex and Genotype Modulate the Dendritic Effects of Developmental Exposure to a Human-Relevant Polychlorinated Biphenyls Mixture in the Juvenile Mouse. Front Neurosci 2021; 15:766802. [PMID: 34924936 PMCID: PMC8678536 DOI: 10.3389/fnins.2021.766802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022] Open
Abstract
While many neurodevelopmental disorders (NDDs) are thought to result from interactions between environmental and genetic risk factors, the identification of specific gene-environment interactions that influence NDD risk remains a critical data gap. We tested the hypothesis that polychlorinated biphenyls (PCBs) interact with human mutations that alter the fidelity of neuronal Ca2+ signaling to confer NDD risk. To test this, we used three transgenic mouse lines that expressed human mutations known to alter Ca2+ signals in neurons: (1) gain-of-function mutation in ryanodine receptor-1 (T4826I-RYR1); (2) CGG-repeat expansion in the 5′ non-coding portion of the fragile X mental retardation gene 1 (FMR1); and (3) a double mutant (DM) that expressed both mutations. Transgenic and wildtype (WT) mice were exposed throughout gestation and lactation to the MARBLES PCB mix at 0.1, 1, or 6 mg/kg in the maternal diet. The MARBLES mix simulates the relative proportions of the twelve most abundant PCB congeners found in serum from pregnant women at increased risk for having a child with an NDD. Using Golgi staining, the effect of developmental PCB exposure on dendritic arborization of pyramidal neurons in the CA1 hippocampus and somatosensory cortex of male and female WT mice was compared to pyramidal neurons from transgenic mice. A multilevel linear mixed-effects model identified a main effect of dose driven by increased dendritic arborization of cortical neurons in the 1 mg/kg PCB dose group. Subsequent analyses with genotypes indicated that the MARBLES PCB mixture had no effect on the dendritic arborization of hippocampal neurons in WT mice of either sex, but significantly increased dendritic arborization of cortical neurons of WT males in the 6 mg/kg PCB dose group. Transgene expression increased sensitivity to the impact of developmental PCB exposure on dendritic arborization in a sex-, and brain region-dependent manner. In conclusion, developmental exposure to PCBs present in the gestational environment of at-risk humans interfered with normal dendritic morphogenesis in the developing mouse brain in a sex-, genotype- and brain region-dependent manner. Overall, these observations provide proof-of-principle evidence that PCBs interact with heritable mutations to modulate a neurodevelopmental outcome of relevance to NDDs.
Collapse
Affiliation(s)
- Kimberly P Keil Stietz
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Carolyn R Klocke
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Tryssa E de Ruyter
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Machelle D Wilson
- Clinical and Translational Science Center, Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
29
|
Leys F, Wenning GK, Fanciulli A. The role of cardiovascular autonomic failure in the differential diagnosis of α-synucleinopathies. Neurol Sci 2021; 43:187-198. [PMID: 34817726 PMCID: PMC8724069 DOI: 10.1007/s10072-021-05746-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022]
Abstract
The α-synucleinopathies comprise a group of adult-onset neurodegenerative disorders including Parkinson’s disease (PD), multiple system atrophy (MSA), dementia with Lewy bodies (DLB,) and — as a restricted non-motor form — pure autonomic failure (PAF). Neuropathologically, the α-synucleinopathies are characterized by aggregates of misfolded α-synuclein in the central and peripheral nervous system. Cardiovascular autonomic failure is a common non-motor symptom in people with PD, a key diagnostic criterion in MSA, a supportive feature for the diagnosis of DLB and disease-defining in PAF. The site of autonomic nervous system lesion differs between the α-synucleinopathies, with a predominantly central lesion pattern in MSA versus a peripheral one in PD, DLB, and PAF. In clinical practice, overlapping autonomic features often challenge the differential diagnosis among the α-synucleinopathies, but also distinguish them from related disorders, such as the tauopathies or other neurodegenerative ataxias. In this review, we discuss the differential diagnostic yield of cardiovascular autonomic failure in individuals presenting with isolated autonomic failure, parkinsonism, cognitive impairment, or cerebellar ataxia.
Collapse
Affiliation(s)
- Fabian Leys
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Gregor K Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Alessandra Fanciulli
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria.
| |
Collapse
|
30
|
Allen EG, Charen K, Hipp HS, Shubeck L, Amin A, He W, Hunter JE, Shelly KE, Sherman SL. Predictors of Comorbid Conditions in Women Who Carry an FMR1 Premutation. Front Psychiatry 2021; 12:715922. [PMID: 34658954 PMCID: PMC8517131 DOI: 10.3389/fpsyt.2021.715922] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose: Women who carry an FMR1 premutation (PM) can experience two well-established PM-associated disorders: fragile X-associated primary ovarian insufficiency (FXPOI, affects ~20-30% carriers) and fragile X-associated tremor-ataxia syndrome (FXTAS, affects ~6-15% carriers); however, emerging evidence indicates that some of these women experience complex health profiles beyond FXPOI and FXTAS. Methods: In an effort to better understand predictors for these comorbid conditions, we collected self-reported medical histories on 413 women who carry an FMR1 PM. Results: There were 22 health conditions reported by at least 9% of women. In an exploratory analysis, 12 variables were tested in logistic regression models for each comorbid condition, including demographic variables, environmental variables, PM-associated factors, and endorsement of depression and/or anxiety. More than half of the comorbid conditions studied were associated with women who self-reported having anxiety. Age, smoking, body mass index (BMI), and depression were also significant predictor variables for specific comorbid conditions. Conclusions: Age, smoking, and BMI were significantly associated with a subset of the comorbid conditions analyzed. Importantly, depression or anxiety were also significantly associated with many of the comorbid health conditions. This work highlights some of the modifiable factors associated with complex health profiles among women with an FMR1 PM.
Collapse
Affiliation(s)
- Emily Graves Allen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Krista Charen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Heather S. Hipp
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Lisa Shubeck
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ashima Amin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Weiya He
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Jessica Ezzell Hunter
- Genomics, Ethics, and Translational Research Program, RTI International, Triangle Park, NC, United States
| | - Katharine E. Shelly
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Stephanie L. Sherman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
31
|
Dulman RS, Auta J, Wandling GM, Patwell R, Zhang H, Pandey SC. Persistence of cerebellar ataxia during chronic ethanol exposure is associated with epigenetic up-regulation of Fmr1 gene expression in rat cerebellum. Alcohol Clin Exp Res 2021; 45:2006-2016. [PMID: 34453331 PMCID: PMC8602769 DOI: 10.1111/acer.14691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Alcohol intoxication produces ataxia by affecting the cerebellum, which coordinates movements. Fragile X mental retardation (FMR) protein is a complex regulator of RNA and synaptic plasticity implicated in fragile X-associated tremor/ataxia syndrome, which features ataxia and increased Fmr1 mRNA expression resulting from epigenetic dysregulation of FMRP. We recently demonstrated that acute ethanol-induced ataxia is associated with increased cerebellar Fmr1 gene expression via histone modifications in rats, but it is unknown whether similar behavioral and molecular changes occur following chronic ethanol exposure. Here, we investigated the effects of chronic ethanol exposure on ataxia and epigenetically regulated changes in Fmr1 expression in the cerebellum. METHODS Male adult Sprague-Dawley rats were trained on the accelerating rotarod and then fed with chronic ethanol or a control Lieber-DeCarli diet while undergoing periodic behavioral testing for ataxia during ethanol exposure and withdrawal. Cerebellar tissues were analyzed for expression of the Fmr1 gene and its targets using a real-time quantitative polymerase chain reaction assay. The epigenetic regulation of Fmr1 was also investigated using a chromatin immunoprecipitation assay. RESULTS Ataxic behavior measured by the accelerating rotarod behavioral test developed during chronic ethanol treatment and persisted at both the 8-h and 24-h withdrawal time points compared to control diet-fed rats. In addition, chronic ethanol treatment resulted in up-regulated expression of Fmr1 mRNA and increased activating epigenetic marks H3K27 acetylation and H3K4 trimethylation at 2 sites within the Fmr1 promoter. Finally, measurement of the expression of relevant FMRP mRNA targets in the cerebellum showed that chronic ethanol up-regulated cAMP response element binding (CREB) Creb1, Psd95, Grm5, and Grin2b mRNA expression without altering Grin2a, Eaa1, or histone acetyltransferases CREB binding protein (Cbp) or p300 mRNA transcripts. CONCLUSIONS These results suggest that epigenetic regulation of Fmr1 and subsequent FMRP regulation of target mRNA transcripts constitute neuroadaptations in the cerebellum that may underlie the persistence of ataxic behavior during chronic ethanol exposure and withdrawal.
Collapse
Affiliation(s)
- Russell S. Dulman
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - James Auta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - Gabriela M. Wandling
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - Ryan Patwell
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, 60612 USA
| | - Subhash C. Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, 60612 USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, 60612 USA
| |
Collapse
|
32
|
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: 13] [Impact Index Per Article: 3.3] [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
|
33
|
Proteau-Lemieux M, Lacroix A, Galarneau L, Corbin F, Lepage JF, Çaku A. The safety and efficacy of metformin in fragile X syndrome: An open-label study. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110307. [PMID: 33757860 DOI: 10.1016/j.pnpbp.2021.110307] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/03/2021] [Accepted: 03/16/2021] [Indexed: 11/18/2022]
Abstract
Fragile X syndrome (FXS) is a rare genetic disorder characterized by a deficit of the fragile X mental retardation protein (FMRP), encoded by the fragile X mental retardation gene (FMR1) on the X chromosome. It has been hypothesized that the absence of FRMP leads to higher levels of Insulin-like Growth Factor 1 (IGF-1) in the brain, possibly contributing to the intellectual impairment characteristic of the disorder. Preclinical studies have shown that metformin downregulates the insulin/IGF-1 signaling pathway, corrects dendritic defects, and improves repetitive behavior in Fmr1 knockout mice. Here, we conducted an open-label study to evaluate: (1) the safety of metformin in normoglycemic individuals with FXS; and (2) the efficacy of metformin to improve aberrant behavior, attention, and to modulate cortical functioning. Fifteen patients with FXS, aged from 17 to 44, received 500 mg of metformin twice/daily over a 9-week treatment period. The primary outcome measures were: (1) the incidence of adverse events (AE); (2) the decrease in IGF-1 levels; and (3) the global score of the Aberrant Behavior Checklist-Community, Fragile X. The secondary outcomes were: (1) the Test of Attentional Performance for children (KiTAP); and (2) the Transcranial Magnetic Stimulation (TMS) parameters measuring cortical excitability. The metformin treatment was well tolerated, with no significant related AE. The TMS data showed an increase in corticospinal inhibition mediated by GABAA and GABAB mechanisms. This study demonstrates the safety of metformin in normoglycemic patients with FXS, and suggests the potential of this medication in modifying GABA-mediated inhibition, a hallmark of FXS pathophysiology. Implications for future clinical trials are discussed.
Collapse
Affiliation(s)
- Mélodie Proteau-Lemieux
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Angélina Lacroix
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Pharmacology, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Luc Galarneau
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Biochemistry, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - François Corbin
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Biochemistry, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Jean-François Lepage
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada.
| | - Artuela Çaku
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Biochemistry, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| |
Collapse
|
34
|
Maltman N, Guilfoyle J, Nayar K, Martin GE, Winston M, Lau JCY, Bush L, Patel S, Lee M, Sideris J, Hall DA, Zhou L, Sharp K, Berry-Kravis E, Losh M. The Phenotypic Profile Associated With the FMR1 Premutation in Women: An Investigation of Clinical-Behavioral, Social-Cognitive, and Executive Abilities. Front Psychiatry 2021; 12:718485. [PMID: 34421690 PMCID: PMC8377357 DOI: 10.3389/fpsyt.2021.718485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/12/2021] [Indexed: 11/23/2022] Open
Abstract
The FMR1 gene in its premutation (PM) state has been linked to a range of clinical and subclinical phenotypes among FMR1 PM carriers, including some subclinical traits associated with autism spectrum disorder (ASD). This study attempted to further characterize the phenotypic profile associated with the FMR1 PM by studying a battery of assessments examining clinical-behavioral traits, social-cognitive, and executive abilities in women carrying the FMR1 PM, and associations with FMR1-related variability. Participants included 152 female FMR1 PM carriers and 75 female controls who were similar in age and IQ, and screened for neuromotor impairments or signs of fragile X-associated tremor/ataxia syndrome. The phenotypic battery included assessments of ASD-related personality and language (i.e., pragmatic) traits, symptoms of anxiety and depression, four different social-cognitive tasks that tapped the ability to read internal states and emotions based on different cues (e.g., facial expressions, biological motion, and complex social scenes), and a measure of executive function. Results revealed a complex phenotypic profile among the PM carrier group, where subtle differences were observed in pragmatic language, executive function, and social-cognitive tasks that involved evaluating basic emotions and trustworthiness. The PM carrier group also showed elevated rates of ASD-related personality traits. In contrast, PM carriers performed similarly to controls on social-cognitive tasks that involved reliance on faces and biological motion. The PM group did not differ from controls on self-reported depression or anxiety symptoms. Using latent profile analysis, we observed three distinct subgroups of PM carriers who varied considerably in their performance across tasks. Among PM carriers, CGG repeat length was a significant predictor of pragmatic language violations. Results suggest a nuanced phenotypic profile characterized by subtle differences in select clinical-behavioral, social-cognitive, and executive abilities associated with the FMR1 PM in women.
Collapse
Affiliation(s)
- Nell Maltman
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Janna Guilfoyle
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Kritika Nayar
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Gary E. Martin
- Department of Communication Sciences and Disorders, St. John's University, Staten Island, NY, United States
| | - Molly Winston
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Joseph C. Y. Lau
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Lauren Bush
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Shivani Patel
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Michelle Lee
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - John Sideris
- Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, United States
| | - Deborah A. Hall
- Department of Neurological Sciences, Rush University, Chicago, IL, United States
| | - Lili Zhou
- Rush University Medical Center, Chicago, IL, United States
| | - Kevin Sharp
- Rush University Medical Center, Chicago, IL, United States
| | | | - Molly Losh
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| |
Collapse
|
35
|
Hong J, Kapoor A, DaWalt LS, Maltman N, Kim B, Berry- Kravis EM, Almeida D, Coe C, Mailick M. Stress and genetics influence hair cortisol in FMR1 premutation carrier mothers of children with fragile X syndrome. Psychoneuroendocrinology 2021; 129:105266. [PMID: 34020265 PMCID: PMC8217368 DOI: 10.1016/j.psyneuen.2021.105266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 01/20/2023]
Abstract
To investigate genetic and environmental influences on cortisol levels, mothers of children with fragile X syndrome (FXS) were studied four times over a 7.5-year period. All participants (n = 84) were carriers of the FMR1 "premutation", a genetic condition associated with impaired HPA axis functioning. Genetic variation was indicated by expansions in the number of CGG (cytosine-guanine-guanine) repeats in the FMR1 gene (67-138 repeats in the present sample). The environmental factor was cumulative exposure to adverse life events during the study period. Cortisol was measured at the beginning of the study via saliva samples and at the end of the study via hair samples; hormone values from these two specimen types were significantly correlated. The interactions between CGG repeat number and adverse life events significantly predicted hair cortisol concentration, including after accounting for the initial salivary cortisol level. For those with fewer CGG repeats, stress exposure was associated with elevated cortisol, the expected response to stress, although women with a higher number of CGGs had a reduced cortisol response to adverse events, which might be related to HPA dysfunction. These results indicate that both exogenous and endogenous factors affect HPA functioning in this population of women.
Collapse
Affiliation(s)
- Jinkuk Hong
- Waisman Center, University of Wisconsin-Madison, United States.
| | - Amita Kapoor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison
| | | | | | - Bryan Kim
- Waisman Center, University of Wisconsin-Madison
| | | | - David Almeida
- Department of Human Development and Family Studies, Pennsylvania State University
| | | | | |
Collapse
|
36
|
Tassanakijpanich N, McKenzie FJ, McLennan YA, Makhoul E, Tassone F, Jasoliya MJ, Romney C, Petrasic IC, Napalinga K, Buchanan CB, Hagerman P, Hagerman R, Casanova EL. Hypermobile Ehlers-Danlos syndrome (hEDS) phenotype in fragile X premutation carriers: case series. J Med Genet 2021; 59:687-690. [PMID: 34193467 DOI: 10.1136/jmedgenet-2020-107609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 06/08/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND While an association between full mutation CGG-repeat expansions of the Fragile X Mental Retardation 1 (FMR1) gene and connective tissue problems are clearly described, problems in fragile X premutation carriers (fXPCs) CGG-repeat range (55-200 repeats) of the FMR1 gene may be overlooked. OBJECTIVE To report five FMR1 fXPCs cases with the hypermobile Ehlers-Danlos syndrome (hEDS) phenotype. METHODS We collected medical histories and FMR1 molecular measures from five cases who presented with joint hypermobility and loose connective tissue and met inclusion criteria for hEDS. RESULTS Five cases were female and ranged between 16 and 49 years. The range of CGG-repeat allele sizes ranged from 66 to 150 repeats. All had symptoms of hEDS since early childhood. Commonalities in molecular pathogenesis and coexisting conditions between the fXPCs and hEDS are also presented. The premutation can lead to a reduction of fragile X mental retardation protein, which is crucial in maintaining functions of the extracellular matrix-related proteins, particularly matrix metallopeptidase 9 and elastin. Moreover, elevated FMR1 messenger RNA causes sequestration of proteins, which results in RNA toxicity. CONCLUSION Both hEDS phenotype and premutation involvement may co-occur because of related commonalities in pathogenesis.
Collapse
Affiliation(s)
- Nattaporn Tassanakijpanich
- Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand.,UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA
| | - Forrest J McKenzie
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Yingratana A McLennan
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Elisabeth Makhoul
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Flora Tassone
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Mittal J Jasoliya
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | | | - Ignacio Cortina Petrasic
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Kaye Napalinga
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,MedMom Institute for Human Development, Pasig City, Philippines
| | | | - Paul Hagerman
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Randi Hagerman
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA .,Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Emily L Casanova
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, South Carolina, USA
| |
Collapse
|
37
|
Mailick MR, Hong J, Movaghar A, DaWalt L, Berry-Kravis EM, Brilliant MH, Boero J, Todd PK, Hall D. Mild Neurological Signs in FMR1 Premutation Women in an Unselected Community-Based Cohort. Mov Disord 2021; 36:2378-2386. [PMID: 34117786 DOI: 10.1002/mds.28683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 05/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Premutation-sized (55-200) CGG repeat expansions in the FMR1 gene cause fragile X-associated tremor/ataxia syndrome (FXTAS). Most studies of premutation carriers utilized reverse ascertainment to identify patients, leading to a selection bias for larger repeats. As shorter CGG premutation repeats are common in the population, understanding their impact on health outcomes has a potentially large public health footprint. OBJECTIVE The study's objective was to compare an unselected group of premutation carriers (n = 35, 55-101 CGG repeats) with matched controls (n = 61, 29-39 CGG repeats) with respect to FXTAS-type signs using structured neurological assessments. METHODS Three neurologists independently rated signs, using an adapted version of the FXTAS Rating Scale (Leehey MA, Berry-Kravis E, Goetz CG, et al. FMR1 CGG repeat length predicts motor dysfunction in premutation carriers. Neurology. 2008). This was a double-blind study, as genetic status (premutation vs. control) was known neither by the participants nor by any of the neurologists. Analyses controlled potentially confounding comorbid conditions in the electronic health record (eg, osteoarthritis and stroke) and probed the association of age with signs. RESULTS Although there was no overall difference between carriers and controls, among individuals without any potentially confounding comorbid diagnoses, there was a statistically significant age-associated elevation in FXTAS-type signs in premutation carriers compared to controls. CONCLUSIONS Among those who do not have other comorbid diagnoses, women who have CGG repeats at the lower end of the premutation range may be at greater risk for ataxia and parkinsonism than their age peers, although their overall risk of developing such clinical features is low. This study should provide reassurance to those who share characteristics with the present cohort. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marsha R Mailick
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jinkuk Hong
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Arezoo Movaghar
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Leann DaWalt
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Murray H Brilliant
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Marshfield Clinic Research Institute, Marshfield, Wisconsin, USA
| | - Jaime Boero
- Marshfield Clinic Research Institute, Marshfield, Wisconsin, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Ann Arbor Veterans Administration Healthcare Center, Ann Arbor, Michigan, USA
| | - Deborah Hall
- Department of Neurological Sciences, Rush University, Chicago, Illinois, USA
| |
Collapse
|
38
|
Storey E, Bui MQ, Stimpson P, Tassone F, Atkinson A, Loesch DZ. Relationships between motor scores and cognitive functioning in FMR1 female premutation X carriers indicate early involvement of cerebello-cerebral pathways. CEREBELLUM & ATAXIAS 2021; 8:15. [PMID: 34116720 PMCID: PMC8196444 DOI: 10.1186/s40673-021-00138-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/28/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Smaller expansions of CGG trinucleotide repeats in the FMR1 X-linked gene termed 'premutation' lead to a neurodegenerative disorder: Fragile X Associated Tremor/Ataxia Syndrome (FXTAS) in nearly half of aged carrier males, and 8-16% females. Core features include intention tremor, ataxia, and cognitive decline, and white matter lesions especially in cerebellar and periventricular locations. A 'toxic' role of elevated and expanded FMR1 mRNA has been linked to the pathogenesis of this disorder. The emerging issue concerns the trajectory of the neurodegenerative changes: is the pathogenetic effect confined to overt clinical manifestations? Here we explore the relationships between motor and cognitive scale scores in a sample of 57 asymptomatic adult female premutation carriers of broad age range. METHODS Three motor scale scores (ICARS-for tremor/ataxia, UPDRS-for parkinsonism, and Clinical Tremor) were related to 11 cognitive tests using Spearman's rank correlations. Robust regression, applied in relationships between all phenotypic measures, and genetic molecular and demographic data, identified age and educational levels as common correlates of these measures, which were then incorporated as confounders in correlation analysis. RESULTS Cognitive tests demonstrating significant correlations with motor scores were those assessing non-verbal reasoning on Matrix Reasoning (p-values from 0.006 to 0.011), and sequencing and alteration on Trails-B (p-values from 0.008 to 0.001). Those showing significant correlations with two motor scores-ICARS and Clinical Tremor- were psychomotor speed on Symbol Digit Modalities (p-values from 0.014 to 0.02) and working memory on Digit Span Backwards (p-values from 0.024 to 0.011). CONCLUSIONS Subtle motor impairments correlating with cognitive, particularly executive, deficits may occur in female premutation carriers not meeting diagnostic criteria for FXTAS. This pattern of cognitive deficits is consistent with those seen in other cerebellar disorders. Our results provide evidence that more than one category of clinical manifestation reflecting cerebellar changes - motor and cognitive - may be simultaneously affected by premutation carriage across a broad age range in asymptomatic carriers.
Collapse
Affiliation(s)
- Elsdon Storey
- Department of Medicine (Neuroscience), Monash University, 5th Floor, Centre Block, Alfred Hospital Campus, Commercial Road, Melbourne, Victoria, 3004, Australia.
| | - Minh Q Bui
- Centre for Molecular, Environmental, Genetic and Analytic, Epidemiology, University of Melbourne, Parkville, Victoria, Australia
| | - Paige Stimpson
- Wellness and Recovery Centre, Monash Medical Centre, Clayton, Victoria, Australia
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine and M.I.N.D. Institute, University of California Davis Medical Center, Davis, California, USA
| | - Anna Atkinson
- School of Psychology and Public Health, La Trobe University, Melbourne, Bundoora, Victoria, Australia
| | - Danuta Z Loesch
- School of Psychology and Public Health, La Trobe University, Melbourne, Bundoora, Victoria, Australia
| |
Collapse
|
39
|
Tokumaru AM, Saito Y, Murayma S. Diffusion-Weighted Imaging is Key to Diagnosing Specific Diseases. Magn Reson Imaging Clin N Am 2021; 29:163-183. [PMID: 33902901 DOI: 10.1016/j.mric.2021.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This article reviews diseases for which persistent signal abnormalities on diffusion-weighted imaging are the key to their diagnosis. Specifically, updated knowledge regarding the neuroimaging patterns of the following diseases is summarized: sporadic Creutzfeldt-Jakob disease, neuronal intranuclear inclusion disease, and hereditary diffuse leukoencephalopathy with axonal spheroids-colony-stimulating factor receptors/adult-onset leukoencephalopathy with axonal spheroids and pigmented glia. In addition, their differential diagnoses; clinical manifestations; and pathologic, genetic, and imaging correlates are discussed.
Collapse
Affiliation(s)
- Aya Midori Tokumaru
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan.
| | - Yuko Saito
- Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Shigeo Murayma
- Brain Bank for Aging Research, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan; Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, 2-2, Yamadaoka, Suita-shi, Osaka-fu 565-0871, Japan
| |
Collapse
|
40
|
Gläser D, Deutsche Gesellschaft Für Humangenetik E V, Berufsverband Deutscher Humangenetiker E V, Hinderhofer K. Leitlinien zur molekulargenetischen Diagnostik: Fragiles-X-Syndrom und andere FMR1-assoziierte Syndrome. MED GENET-BERLIN 2021; 33:65-73. [PMID: 38836214 PMCID: PMC11006362 DOI: 10.1515/medgen-2021-2059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Affiliation(s)
- Dieter Gläser
- Genetikum, Wegenerstr. 15, D-89231Neu-Ulm, Deutschland
| | - Deutsche Gesellschaft Für Humangenetik E V
- Genetikum, Wegenerstr. 15, D-89231Neu-Ulm, Deutschland
- Universitätsklinikum Heidelberg, Institut für Humangenetik, Im Neuenheimer Feld 366, D-69120Heidelberg, Deutschland
| | - Berufsverband Deutscher Humangenetiker E V
- Genetikum, Wegenerstr. 15, D-89231Neu-Ulm, Deutschland
- Universitätsklinikum Heidelberg, Institut für Humangenetik, Im Neuenheimer Feld 366, D-69120Heidelberg, Deutschland
| | - Katrin Hinderhofer
- Universitätsklinikum Heidelberg, Institut für Humangenetik, Im Neuenheimer Feld 366, D-69120Heidelberg, Deutschland
| |
Collapse
|
41
|
'Essential Tremor' Phenotype in FMR1 Premutation/Gray Zone Sibling Series: Exploring Possible Genetic Modifiers. Twin Res Hum Genet 2021; 24:95-102. [PMID: 33757613 DOI: 10.1017/thg.2021.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) occurs in carriers of fragile X mental retardation 1 (FMR1) X-linked small CGG expansion (gray zone [GZ] and premutation [PM]) alleles, containing 41-200 repeats. Major features comprise kinetic tremor, gait ataxia, cognitive decline and cerebellar peduncular white matter lesions, but atypical/incomplete FXTAS may occur. We explored the possibility of polygenic effects modifying the FXTAS spectrum phenotypes. We used three motor scales and selected cognitive tests in a series of three males and three females from a single sibship carrying PM or GZ alleles (44 to 75 repeats). The molecular profiles from these siblings were determined by genomewide association study with single-nucleotide polymorphism (SNP) genotyping by Illumina Global Screening Array. Nonparametric linkage analysis was applied and Parkinson's disease (PD) polygenic risk scores (PRSs) were calculated for all the siblings, based on 107 known risk variants. All male and female siblings manifested similar kinetic tremor phenotypes. In contrast to FXTAS, they showed negligible gait ataxia, and few white matter lesions on MRI. Cognitive functioning was unaffected. Suggestive evidence of linkage to a broad region of the short arm of chromosome 10 was obtained, and median PD PRS for the sibship fell within the top 30% of a sample of over 500,000 UK and Australian controls. The genomewide study results are suggestive of modifying effects of genetic risk loci linked to PD, on the neurological phenotype of FMR1-CGG small expansion carriers, resulting in an oligosymptomatic kinetic tremor seen in FXTAS spectrum, but also consistent with essential tremor.
Collapse
|
42
|
Tassanakijpanich N, Hagerman RJ, Worachotekamjorn J. Fragile X premutation and associated health conditions: A review. Clin Genet 2021; 99:751-760. [PMID: 33443313 DOI: 10.1111/cge.13924] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/17/2022]
Abstract
Fragile X syndrome (FXS) is the most common single gene disorder, which causes autism and intellectual disability. The fragile X mental retardation 1 (FMR1) gene is silenced when cytosine-guanine-guanine (CGG) triplet repeats exceed 200, which is the full mutation that causes FXS. Carriers of FXS have a CGG repeat between 55 and 200, which is defined as a premutation and transcription of the gene is overactive with high levels of the FMR1 mRNA. Most carriers of the premutation have normal levels of fragile X mental retardation protein (FMRP) and a normal intelligence, but in the upper range of the premutation (120-200) the FMRP level may be lower than normal. The clinical problems associated with the premutation are caused by the RNA toxicity associated with increased FMR1 mRNA levels, although for some mildly lowered FMRP can cause problems associated with FXS. The RNA toxicity causes various health problems in the carriers including but not limited to fragile X-associated tremor/ataxia syndrome, fragile X-associated primary ovarian insufficiency, and fragile X-associated neuropsychiatric disorders. Since some individuals with neuropsychiatric problems do not meet the severity for a diagnosis of a "disorder" then the condition can be labeled as fragile X premutation associated condition (FXPAC). Physicians must be able to recognize these health problems in the carriers and provide appropriate management.
Collapse
Affiliation(s)
| | - Randi J Hagerman
- UC Davis MIND Institute, UC Davis Health, Sacramento, California, USA.,Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California, USA
| | | |
Collapse
|
43
|
Bredin-Oja SL, Warren SF, Swinburne Romine RE, Fleming KK, Brady N, Berry-Kravis E. Word retrieval difficulty in adult females with the FMR1 premutation: Changes over time and across contexts. Brain Cogn 2021; 148:105694. [PMID: 33503544 DOI: 10.1016/j.bandc.2021.105694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 01/21/2023]
Abstract
Individuals with a premutation of the fragile X mental retardation (FMR1) gene are at risk for a variety of psychological, physical, and cognitive issues, including difficulty with word retrieval. The present study examined three indicators of word retrieval difficulty; reduced productivity, reduced lexical diversity, and increased errors in word retrieval in a group of 38 female premutation carriers during standard-length speech samples collected over a period of eight years. Our results revealed that as women aged, they produced fewer words, produced fewer different words, and had greater word retrieval errors. In addition, the rate of word retrieval errors was highly correlated between two speaking contexts, indicating that this difficulty was pervasive and not solely the result of speaking in monologue. Our results suggest that subtle areas of cognitive decline emerge at a much earlier age among female premutation carriers than would be expected during healthy aging.
Collapse
Affiliation(s)
- Shelley L Bredin-Oja
- Life Span Institute, University of Kansas, 1052 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA.
| | - Steven F Warren
- Life Span Institute, University of Kansas, 1052 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA; Department of Speech-Language-Hearing: Sciences and Disorders, University of Kansas, 3001 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Rebecca E Swinburne Romine
- Life Span Institute, University of Kansas, 1052 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Kandace K Fleming
- Life Span Institute, University of Kansas, 1052 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Nancy Brady
- Life Span Institute, University of Kansas, 1052 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA; Department of Speech-Language-Hearing: Sciences and Disorders, University of Kansas, 3001 Dole Human Development Center, 1000 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Elizbeth Berry-Kravis
- Department of Pediatrics, Neurological Sciences and Biochemistry, Rush University Medical Center, 1725 West Harrison Street, Suite 718, Chicago, IL 60612, USA
| |
Collapse
|
44
|
Loesch DZ, Tassone F, Atkinson A, Stimpson P, Trost N, Pountney DL, Storey E. Differential Progression of Motor Dysfunction Between Male and Female Fragile X Premutation Carriers Reveals Novel Aspects of Sex-Specific Neural Involvement. Front Mol Biosci 2021; 7:577246. [PMID: 33511153 PMCID: PMC7835843 DOI: 10.3389/fmolb.2020.577246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Expansions of the CGG repeat in the non-coding segment of the FMR1 X-linked gene are associated with a variety of phenotypic changes. Large expansions (>200 repeats), which cause a severe neurodevelopmental disorder, the fragile x syndrome (FXS), are transmitted from the mothers carrying smaller, unstable expansions ranging from 55 to 200 repeats, termed the fragile X premutation. Female carriers of this premutation may themselves experience a wide range of clinical problems throughout their lifespan, the most severe being the late onset neurodegenerative condition called "Fragile X-Associated Tremor Ataxia Syndrome" (FXTAS), occurring between 8 and 16% of these carriers. Male premutation carriers, although they do not transmit expanded alleles to their daughters, have a much higher risk (40-50%) of developing FXTAS. Although this disorder is more prevalent and severe in male than female carriers, specific sex differences in clinical manifestations and progress of the FXTAS spectrum have been poorly documented. Here we compare the pattern and rate of progression (per year) in three motor scales including tremor/ataxia (ICARS), tremor (Clinical Tremor Rating scale, CRST), and parkinsonism (UPDRS), and in several cognitive and psychiatric tests scores, between 13 female and 9 male carriers initially having at least one of the motor scores ≥10. Moreover, we document the differences in each of the clinical and cognitive measures between the cross-sectional samples of 21 female and 24 male premutation carriers of comparable ages with FXTAS spectrum disorder (FSD), that is, who manifest one or more features of FXTAS. The results of progression assessment showed that it was more than twice the rate in male than in female carriers for the ICARS-both gait ataxia and kinetic tremor domains and twice as high in males on the CRST scale. In contrast, sex difference was negligible for the rate of progress in UPDRS, and all the cognitive measures. The overall psychiatric pathology score (SCL-90), as well as Anxiety and Obsessive/Compulsive domain scores, showed a significant increase only in the female sample. The pattern of sex differences for progression in motor scores was consistent with the results of comparison between larger, cross-sectional samples of male and female carriers affected with the FSD. These results were in concert with sex-specific distribution of MRI T2 white matter hyperintensities: all males, but no females, showed the middle cerebellar peduncle white matter hyperintensities (MCP sign), although the distribution and severity of these hyperintensities in the other brain regions were not dissimilar between the two sexes. In conclusion, the magnitude and specific pattern of sex differences in manifestations and progression of clinically recorded changes in motor performance and MRI lesion distribution support, on clinical grounds, the possibility of certain sex-limited factor(s) which, beyond the predictable effect of the second, normal FMR1 alleles in female premutation carriers, may have neuroprotective effects, specifically concerning the cerebellar circuitry.
Collapse
Affiliation(s)
- Danuta Z. Loesch
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - 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 Center, Davis, CA, United States
| | - Anna Atkinson
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - Paige Stimpson
- Wellness and Recovery Centre, Monash Medical Centre, Clayton, VIC, Australia
| | - Nicholas Trost
- Medical Imaging Department, St Vincent's Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Dean L. Pountney
- Neurodegeneration Research Group, School of Medical Science, Griffith University, Gold Coast Campus, Southport, NC, Australia
| | - Elsdon Storey
- Department of Medicine (Neuroscience), Monash University, Alfred Hospital Campus, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Shukla T, de la Peña JB, Perish JM, Ploski JE, Stumpf CR, Webster KR, Thorn CA, Campbell ZT. A Highly Selective MNK Inhibitor Rescues Deficits Associated with Fragile X Syndrome in Mice. Neurotherapeutics 2021; 18:624-639. [PMID: 33006091 PMCID: PMC8116363 DOI: 10.1007/s13311-020-00932-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2020] [Indexed: 12/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited source of intellectual disability in humans. FXS is caused by mutations that trigger epigenetic silencing of the Fmr1 gene. Loss of Fmr1 results in increased activity of the mitogen-activated protein kinase (MAPK) pathway. An important downstream consequence is activation of the mitogen-activated protein kinase interacting protein kinase (MNK). MNK phosphorylates the mRNA cap-binding protein, eukaryotic initiation factor 4E (eIF4E). Excessive phosphorylation of eIF4E has been directly implicated in the cognitive and behavioral deficits associated with FXS. Pharmacological reduction of eIF4E phosphorylation is one potential strategy for FXS treatment. We demonstrate that systemic dosing of a highly specific, orally available MNK inhibitor, eFT508, attenuates numerous deficits associated with loss of Fmr1 in mice. eFT508 resolves a range of phenotypic abnormalities associated with FXS including macroorchidism, aberrant spinogenesis, and alterations in synaptic plasticity. Key behavioral deficits related to anxiety, social interaction, obsessive and repetitive activities, and object recognition are ameliorated by eFT508. Collectively, this work establishes eFT508 as a potential means to reverse deficits associated with FXS.
Collapse
Affiliation(s)
- Tarjani Shukla
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - John M Perish
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | | | | | - Catherine A Thorn
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA.
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
46
|
Elias-Mas A, Alvarez-Mora MI, Caro-Benito C, Rodriguez-Revenga L. Neuroimaging Insight Into Fragile X-Associated Neuropsychiatric Disorders: Literature Review. Front Psychiatry 2021; 12:728952. [PMID: 34721105 PMCID: PMC8554234 DOI: 10.3389/fpsyt.2021.728952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022] Open
Abstract
FMR1 premutation is defined by 55-200 CGG repeats in the Fragile X Mental Retardation 1 (FMR1) gene. FMR1 premutation carriers are at risk of developing a neurodegenerative disease called fragile X-associated tremor/ataxia syndrome (FXTAS) and Fragile X-associated primary ovarian insufficiency (FXPOI) in adulthood. In the last years an increasingly board spectrum of clinical manifestations including psychiatric disorders have been described as occurring at a greater frequency among FMR1 premutation carriers. Herein, we reviewed the neuroimaging findings reported in relation with psychiatric symptomatology in adult FMR1 premutation carriers. A structured electronic literature search was conducted on FMR1 premutation and neuroimaging yielding a total of 3,229 articles examined. Of these, 7 articles were analyzed and are included in this review. The results showed that the main radiological findings among adult FMR1 premutation carriers presenting neuropsychiatric disorders were found on the amygdala and hippocampus, being the functional abnormalities more consistent and the volumetric changes more inconsistent among studies. From a molecular perspective, CGG repeat size, FMR1 mRNA and FMRP levels have been investigated in relation with the neuroimaging findings. Based on the published results, FMRP might play a key role in the pathophysiology of the psychiatric symptoms described among FMR1 premutation carriers. However, additional studies including further probes of brain function and a broader scope of psychiatric symptom measurement are required in order to obtain a comprehensive landscape of the neuropsychiatric phenotype associated with the FMR1 premutation.
Collapse
Affiliation(s)
- Andrea Elias-Mas
- Radiology Department, Hospital Universitari Mútua de Terrassa, Terrassa, Spain.,Institute for Research and Innovation Parc Taulí (I3PT), Sabadell, Spain.,Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Maria Isabel Alvarez-Mora
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Laia Rodriguez-Revenga
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
47
|
Loesch DZ, Kemp BE, Bui MQ, Fisher PR, Allan CY, Sanislav O, Ngoei KRW, Atkinson A, Tassone F, Annesley SJ, Storey E. Cellular Bioenergetics and AMPK and TORC1 Signalling in Blood Lymphoblasts Are Biomarkers of Clinical Status in FMR1 Premutation Carriers. Front Psychiatry 2021; 12:747268. [PMID: 34880790 PMCID: PMC8645580 DOI: 10.3389/fpsyt.2021.747268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
Fragile X Associated Tremor/Ataxia Syndrome (FXTAS) is a neurodegenerative disorder affecting carriers of premutation alleles (PM) of the X-linked FMR1 gene, which contain CGG repeat expansions of 55-200 range in a non-coding region. This late-onset disorder is characterised by the presence of tremor/ataxia and cognitive decline, associated with the white matter lesions throughout the brain, especially involving the middle cerebellar peduncles. Nearly half of older male and ~ 20% of female PM carriers develop FXTAS. While there is evidence for mitochondrial dysfunction in neural and some peripheral tissues from FXTAS patients (though less obvious in the non-FXTAS PM carriers), the results from peripheral blood mononuclear cells (PBMC) are still controversial. Motor, cognitive, and neuropsychiatric impairments were correlated with measures of mitochondrial and non-mitochondrial respiratory activity, AMPK, and TORC1 cellular stress-sensing protein kinases, and CGG repeat size, in a sample of adult FXTAS male and female carriers. Moreover, the levels of these cellular measures, all derived from Epstein- Barr virus (EBV)- transformed and easily accessible blood lymphoblasts, were compared between the FXTAS (N = 23) and non-FXTAS (n = 30) subgroups, and with baseline data from 33 healthy non-carriers. A significant hyperactivity of cellular bioenergetics components as compared with the baseline data, more marked in the non-FXTAS PMs, was negatively correlated with repeat numbers at the lower end of the CGG-PM distribution. Significant associations of these components with motor impairment measures, including tremor-ataxia and parkinsonism, and neuropsychiatric changes, were prevalent in the FXTAS subgroup. Moreover, a striking elevation of AMPK activity, and a decrease in TORC1 levels, especially in the non-FXTAS carriers, were related to the size of CGG expansion. The bioenergetics changes in blood lymphoblasts are biomarkers of the clinical status of FMR1 carriers. The relationship between these changes and neurological involvement in the affected carriers suggests that brain bioenergetic alterations are reflected in this peripheral tissue. A possible neuroprotective role of stress sensing kinase, AMPK, in PM carriers, should be addressed in future longitudinal studies. A decreased level of TORC1-the mechanistic target of the rapamycin complex, suggests a possible future approach to therapy in FXTAS.
Collapse
Affiliation(s)
- Danuta Z Loesch
- School of Psychology and Public Health, La Trobe University, Bundoora, VA, Australia
| | - Bruce E Kemp
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VA, Australia.,St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, VA, Australia
| | - Minh Q Bui
- Centre for Molecular, Environmental, Genetic and Analytic, Epidemiology, University of Melbourne, Parkville, VA, Australia
| | - Paul R Fisher
- Department of Physiology Anatomy and Microbiology, La Trobe University, Bundoora, VA, Australia
| | - Claire Y Allan
- Department of Physiology Anatomy and Microbiology, La Trobe University, Bundoora, VA, Australia
| | - Oana Sanislav
- Department of Physiology Anatomy and Microbiology, La Trobe University, Bundoora, VA, Australia
| | - Kevin R W Ngoei
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, VA, Australia
| | - Anna Atkinson
- School of Psychology and Public Health, La Trobe University, Bundoora, VA, Australia
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States.,Department of Biochemistry and Molecular Medicine M.I.N.D. Institute, University of California Davis Medical Center, Davis, Sacramento, CA, United States
| | - Sarah J Annesley
- Department of Physiology Anatomy and Microbiology, La Trobe University, Bundoora, VA, Australia
| | - Elsdon Storey
- Department of Medicine (Neuroscience), Monash University, Alfred Hospital Campus, Melbourne, VIC, Australia
| |
Collapse
|
48
|
Hocking DR, Loesch DZ, Stimpson P, Tassone F, Atkinson A, Storey E. Delineating the Relationships Between Motor, Cognitive-Executive and Psychiatric Symptoms in Female FMR1 Premutation Carriers. Front Psychiatry 2021; 12:742929. [PMID: 34925088 PMCID: PMC8678043 DOI: 10.3389/fpsyt.2021.742929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Premutation expansions (55-200 CGG repeats) of the Fragile X Mental Retardation 1 (FMR1) gene on the X chromosome are associated with a range of clinical features. Apart from the most severe - Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS) - where the most typical white matter changes affect cerebellar peduncles, more subtle changes may include impairment of executive functioning, affective disorders and/or subtle motor changes. Here we aimed to examine whether performance in selected components of executive functioning is associated with subclinical psychiatric symptoms in non-FXTAS, adult females carrying the FMR1 premutation. Methods and Sample: A total of 47 female premutation carriers (sub-symptomatic for FXTAS) of wide age range (26-77 years; M = 50.3; SD = 10.9) were assessed using standard neuropsychological tests, three motor rating scales and self-reported measures of psychiatric symptoms using the Symptom Checklist-90-Revised (SCL-90-R). Results: After adjusting for age and educational level where appropriate, both non-verbal reasoning and response inhibition as assessed on the Stroop task (i.e., the ability to resolve cognitive interference) were associated with a range of primary psychiatric symptom dimensions, and response inhibition uniquely predicted some primary symptoms and global psychiatric features. Importantly, lower scores (worse performance) in response inhibition were also strongly correlated with higher (worse) scores on standard motor rating scales for tremor-ataxia and for parkinsonism. Conclusion: These results provide evidence for the importance of response inhibition in the manifestation of psychiatric symptoms and subtle tremor-ataxia motor features, suggestive of the presence of early cerebellar changes in female premutation carriers.
Collapse
Affiliation(s)
- Darren R Hocking
- Developmental Neuromotor and Cognition Lab, School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Danuta Z Loesch
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Paige Stimpson
- Psychology Department, Monash Health, Clayton, VIC, Australia
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine and M.I.N.D. Institute, University of California Davis Medical Center, University of California, Davis, Davis, CA, United States
| | - Anna Atkinson
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Elsdon Storey
- Department of Medicine (Neuroscience), Monash University, Alfred Hospital Campus, Melbourne, VIC, Australia
| |
Collapse
|
49
|
Hong J, DaWalt L, Baker MW, Berry-Kravis EM, Mailick MR. Is FMR1 CGG Repeat Number Polymorphism Associated With Phenotypic Variation in the General Population? Report From a Cohort of 5,499 Adults. Front Psychiatry 2021; 12:727085. [PMID: 34456771 PMCID: PMC8385267 DOI: 10.3389/fpsyt.2021.727085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
FMR1 CGG repeat length was assayed in 5499 research participants (2637 men and 2862 women) in the Wisconsin Longitudinal Study (WLS), a population-based cohort. Most past research has focused on clinically-ascertained individuals with expansions in CGG repeats, either those with fragile X syndrome (> 200 CGG repeats), the FMR1 premutation (55-200 repeats), or in the gray zone (variously defined as 45-54 or 41-54 repeats). In contrast, the WLS is a unique source of data that was obtained from an unselected cohort of individuals from the general population for whom FMR1 CGG repeat length was assayed. The WLS is a random sample of one-third of all high school seniors in the state of Wisconsin in 1957. The most recent round of data collection was in 2011; thus, the study spanned over 50 years. Saliva samples were obtained from 69% of surviving members of the cohort in 2008 and 2011, from which CGG repeats were assayed. With one exception, the CGG repeat length of all members of this cohort was below 100 (ranging from 7 to 84). The present study evaluated the genotype-phenotype associations of CGG repeat number and IQ, college graduation, age at menopause, number of biological children, having a child with intellectual or developmental disabilities, and the likelihood of experiencing an episode of depression during adulthood. Linear and curvilinear effects were probed. Although effect sizes were small, significant associations were found between CGG repeat length and high school IQ score, college graduation, number of biological children, age at menopause, and the likelihood of having an episode of depression. However, there was no significant association between repeat length and having a child diagnosed with an IDD condition. This study demonstrates a continuum of phenotype effects with FMR1 repeat lengths and illustrates how research inspired by a rare genetic condition (such as fragile X syndrome) can be used to probe genotype-phenotype associations in the general population.
Collapse
Affiliation(s)
- Jinkuk Hong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Leann DaWalt
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Mei Wang Baker
- Wisconsin State Laboratory of Hygiene, Madison, WI, United States
| | - Elizabeth M Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, IL, United States
| | - Marsha R Mailick
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
50
|
Moser C, Schmitt L, Schmidt J, Fairchild A, Klusek J. Response Inhibition Deficits in Women with the FMR1 Premutation are Associated with Age and Fall Risk. Brain Cogn 2020; 148:105675. [PMID: 33387817 DOI: 10.1016/j.bandc.2020.105675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/04/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022]
Abstract
One in 113-178 females worldwide carry a premutation allele on the FMR1 gene. The FMR1 premutation is linked to neurocognitive and neuromotor impairments, although the phenotype is not fully understood, particularly with respect to age effects. This study sought to define oculomotor response inhibition skills in women with the FMR1 premutation and their association with age and fall risk. We employed an antisaccade eye-tracking paradigm to index oculomotor inhibition skills in 35 women with the FMR1 premutation and 28 control women. The FMR1 premutation group exhibited longer antisaccade latency and reduced accuracy relative to controls, indicating deficient response inhibition skills. Longer response latency was associated with older age in the FMR1 premutation and was also predictive of fall risk. Findings highlight the utility of the antisaccade paradigm for detecting early signs of age-related executive decline in the FMR1 premutation, which is related to fall risk. Findings support the need for clinical prevention efforts to decrease and delay the trajectory of age-related executive decline in women with the FMR1 premutation during midlife.
Collapse
Affiliation(s)
- Carly Moser
- Communication Sciences and Disorders, University of South Carolina, 1705 College Street, Columbia, South Carolina, 29208, USA
| | - Lyndsay Schmitt
- Communication Sciences and Disorders, University of South Carolina, 1705 College Street, Columbia, South Carolina, 29208, USA
| | - Joseph Schmidt
- Department of Psychology, University of Central Florida, 4111 Pictor Lane, Orlando, FL 32816, Orlando, Florida 32816, USA
| | - Amanda Fairchild
- Department of Psychology, University of South Carolina, 1512 Pendleton Street, Columbia, South Carolina, 29208, USA
| | - Jessica Klusek
- Communication Sciences and Disorders, University of South Carolina, 1705 College Street, Columbia, South Carolina, 29208, USA.
| |
Collapse
|