1
|
Khandjian EW, Moss T, Rose TM, Robert C, Davidovic L. The fragile X proteins' enigma: to be or not to be nucleolar. Front Cell Dev Biol 2024; 12:1448209. [PMID: 39156973 PMCID: PMC11327008 DOI: 10.3389/fcell.2024.1448209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Affiliation(s)
- Edouard W. Khandjian
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, et Centre de Recherche Cervo, Québec, QC, Canada
| | - Tom Moss
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, et Centre de Recherche sur le Cancer, Axe Oncologie, Centre de Recherche du CHUQ, Université Laval, Québec, QC, Canada
| | - Timothy M. Rose
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Claude Robert
- Département des Sciences Animales, Université Laval, Québec, QC, Canada
| | - Laetitia Davidovic
- Centre National de la Recherche Scientifique UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Inserm U1318, Université Côte d’Azur, Valbonne, France
| |
Collapse
|
2
|
Sirois CL, Guo Y, Li M, Wolkoff NE, Korabelnikov T, Sandoval S, Lee J, Shen M, Contractor A, Sousa AMM, Bhattacharyya A, Zhao X. CGG repeats in the human FMR1 gene regulate mRNA localization and cellular stress in developing neurons. Cell Rep 2024; 43:114330. [PMID: 38865241 PMCID: PMC11240841 DOI: 10.1016/j.celrep.2024.114330] [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: 11/18/2023] [Revised: 04/18/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
The human genome has many short tandem repeats, yet the normal functions of these repeats are unclear. The 5' untranslated region (UTR) of the fragile X messenger ribonucleoprotein 1 (FMR1) gene contains polymorphic CGG repeats, the length of which has differing effects on FMR1 expression and human health, including the neurodevelopmental disorder fragile X syndrome. We deleted the CGG repeats in the FMR1 gene (0CGG) in human stem cells and examined the effects on differentiated neurons. 0CGG neurons have altered subcellular localization of FMR1 mRNA and protein, and differential expression of cellular stress proteins compared with neurons with normal repeats (31CGG). In addition, 0CGG neurons have altered responses to glucocorticoid receptor (GR) activation, including FMR1 mRNA localization, GR chaperone HSP90α expression, GR localization, and cellular stress protein levels. Therefore, the CGG repeats in the FMR1 gene are important for the homeostatic responses of neurons to stress signals.
Collapse
Affiliation(s)
- Carissa L Sirois
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yu Guo
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Meng Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Natalie E Wolkoff
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Tomer Korabelnikov
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Soraya Sandoval
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jiyoun Lee
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Minjie Shen
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Amaya Contractor
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andre M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
3
|
Ledoux N, Lelong EIJ, Simard A, Hussein S, Adjibade P, Lambert JP, Mazroui R. The Identification of Nuclear FMRP Isoform Iso6 Partners. Cells 2023; 12:2807. [PMID: 38132127 PMCID: PMC10742089 DOI: 10.3390/cells12242807] [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: 10/26/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
A deficiency of FMRP, a canonical RNA-binding protein, causes the development of Fragile X Syndrome (FXS), which is characterised by multiple phenotypes, including neurodevelopmental disorders, intellectual disability, and autism. Due to the alternative splicing of the encoding FMR1 gene, multiple FMRP isoforms are produced consisting of full-length predominantly cytoplasmic (i.e., iso1) isoforms involved in translation and truncated nuclear (i.e., iso6) isoforms with orphan functions. However, we recently implicated nuclear FMRP isoforms in DNA damage response, showing that they negatively regulate the accumulation of anaphase DNA genomic instability bridges. This finding provided evidence that the cytoplasmic and nuclear functions of FMRP are uncoupled played by respective cytoplasmic and nuclear isoforms, potentially involving specific interactions. While interaction partners of cytoplasmic FMRP have been reported, the identity of nuclear FMRP isoform partners remains to be established. Using affinity purification coupled with mass spectrometry, we mapped the nuclear interactome of the FMRP isoform iso6 in U2OS. In doing so, we found FMRP nuclear interaction partners to be involved in RNA processing, pre-mRNA splicing, ribosome biogenesis, DNA replication and damage response, chromatin remodeling and chromosome segregation. By comparing interactions between nuclear iso6 and cytoplasmic iso1, we report a set of partners that bind specifically to the nuclear isoforms, mainly proteins involved in DNA-associated processes and proteasomal proteins, which is consistent with our finding that proteasome targets the nuclear FMRP iso6. The specific interactions with the nuclear isoform 6 are regulated by replication stress, while those with the cytoplasmic isoform 1 are largely insensitive to such stress, further supporting a specific role of nuclear isoforms in DNA damage response induced by replicative stress, potentially regulated by the proteasome.
Collapse
Affiliation(s)
- Nassim Ledoux
- Centre de Recherche du CHU de Québec—Université Laval, Axe Oncologie, Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (N.L.); (E.I.J.L.); (A.S.); (S.H.); (P.A.)
| | - Emeline I. J. Lelong
- Centre de Recherche du CHU de Québec—Université Laval, Axe Oncologie, Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (N.L.); (E.I.J.L.); (A.S.); (S.H.); (P.A.)
| | - Alexandre Simard
- Centre de Recherche du CHU de Québec—Université Laval, Axe Oncologie, Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (N.L.); (E.I.J.L.); (A.S.); (S.H.); (P.A.)
| | - Samer Hussein
- Centre de Recherche du CHU de Québec—Université Laval, Axe Oncologie, Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (N.L.); (E.I.J.L.); (A.S.); (S.H.); (P.A.)
| | - Pauline Adjibade
- Centre de Recherche du CHU de Québec—Université Laval, Axe Oncologie, Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (N.L.); (E.I.J.L.); (A.S.); (S.H.); (P.A.)
| | - Jean-Philippe Lambert
- Centre de Recherche du CHU de Québec—Université Laval, Axe Endocrinologie et néphrologie, Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada;
- PROTEO, Le Regroupement Québécois De Recherche Sur La Fonction, L’ingénierie et Les Applications des Protéines, Université Laval, Québec, QC G1V 0A6, Canada
| | - Rachid Mazroui
- Centre de Recherche du CHU de Québec—Université Laval, Axe Oncologie, Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (N.L.); (E.I.J.L.); (A.S.); (S.H.); (P.A.)
| |
Collapse
|
4
|
Tuncay IO, DeVries D, Gogate A, Kaur K, Kumar A, Xing C, Goodspeed K, Seyoum-Tesfa L, Chahrour MH. The genetics of autism spectrum disorder in an East African familial cohort. CELL GENOMICS 2023; 3:100322. [PMID: 37492102 PMCID: PMC10363748 DOI: 10.1016/j.xgen.2023.100322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/09/2023] [Accepted: 04/16/2023] [Indexed: 07/27/2023]
Abstract
Autism spectrum disorder (ASD) is a group of complex neurodevelopmental conditions affecting communication and social interaction in 2.3% of children. Studies that demonstrated its complex genetic architecture have been mainly performed in populations of European ancestry. We investigate the genetics of ASD in an East African cohort (129 individuals) from a population with higher prevalence (5%). Whole-genome sequencing identified 2.13 million private variants in the cohort and potentially pathogenic variants in known ASD genes (including CACNA1C, CHD7, FMR1, and TCF7L2). Admixture analysis demonstrated that the cohort comprises two ancestral populations, African and Eurasian. Admixture mapping discovered 10 regions that confer ASD risk on the African haplotypes, containing several known ASD genes. The increased ASD prevalence in this population suggests decreased heterogeneity in the underlying genetic etiology, enabling risk allele identification. Our approach emphasizes the power of African genetic variation and admixture analysis to inform the architecture of complex disorders.
Collapse
Affiliation(s)
- Islam Oguz Tuncay
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Darlene DeVries
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashlesha Gogate
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kiran Kaur
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kimberly Goodspeed
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Maria H Chahrour
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
5
|
Gene therapy using human FMRP isoforms driven by the human FMR1 promoter rescues fragile X syndrome mouse deficits. Mol Ther Methods Clin Dev 2022; 27:246-258. [PMID: 36320413 PMCID: PMC9593309 DOI: 10.1016/j.omtm.2022.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022]
Abstract
Fragile X syndrome (FXS) is caused by the loss of the fragile X messenger ribonucleoprotein 1 (FMRP) encoded by the FMR1 gene. Gene therapy using adeno-associated virus (AAV) to restore FMRP expression is a promising therapeutic strategy. However, so far AAV gene therapy tests for FXS only utilized rodent FMRPs driven by promoters other than the human FMR1 promoter. Restoration of human FMRP in appropriate cell types and at physiological levels, preferably driven by the human FMR1 promoter, would be more suitable for its clinical use. Herein, we generated two human FMR1 promoter subdomains that effectively drive gene expression. When AAVs expressing two different human FMRP isoforms under the control of a human FMR1 promoter subdomain were administered into bilateral ventricles of neonatal Fmr1 -/y and wild-type (WT) mice, both human FMRP isoforms were expressed throughout the brain in a pattern reminiscent to that of mouse FMRP. Importantly, human FMRP expression attenuated social behavior deficits and stereotyped and repetitive behavior, and reversed dysmorphological dendritic spines in Fmr1 -/y mice, without affecting WT mouse behaviors. Our results demonstrate that human FMR1 promoter can effectively drive human FMRP expression in the brain to attenuate Fmr1 -/y mouse deficits, strengthening the notion of using AAV gene therapy for FXS treatment.
Collapse
|
6
|
Kieffer F, Hilal F, Gay AS, Debayle D, Pronot M, Poupon G, Lacagne I, Bardoni B, Martin S, Gwizdek C. Combining affinity purification and mass spectrometry to define the network of the nuclear proteins interacting with the N-terminal region of FMRP. Front Mol Biosci 2022; 9:954087. [PMID: 36237573 PMCID: PMC9553004 DOI: 10.3389/fmolb.2022.954087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Fragile X-Syndrome (FXS) represents the most common inherited form of intellectual disability and the leading monogenic cause of Autism Spectrum Disorders. In most cases, this disease results from the absence of expression of the protein FMRP encoded by the FMR1 gene (Fragile X messenger ribonucleoprotein 1). FMRP is mainly defined as a cytoplasmic RNA-binding protein regulating the local translation of thousands of target mRNAs. Interestingly, FMRP is also able to shuttle between the nucleus and the cytoplasm. However, to date, its roles in the nucleus of mammalian neurons are just emerging. To broaden our insight into the contribution of nuclear FMRP in mammalian neuronal physiology, we identified here a nuclear interactome of the protein by combining subcellular fractionation of rat forebrains with pull‐ down affinity purification and mass spectrometry analysis. By this approach, we listed 55 candidate nuclear partners. This interactome includes known nuclear FMRP-binding proteins as Adar or Rbm14 as well as several novel candidates, notably Ddx41, Poldip3, or Hnrnpa3 that we further validated by target‐specific approaches. Through our approach, we identified factors involved in different steps of mRNA biogenesis, as transcription, splicing, editing or nuclear export, revealing a potential central regulatory function of FMRP in the biogenesis of its target mRNAs. Therefore, our work considerably enlarges the nuclear proteins interaction network of FMRP in mammalian neurons and lays the basis for exciting future mechanistic studies deepening the roles of nuclear FMRP in neuronal physiology and the etiology of the FXS.
Collapse
Affiliation(s)
- Félicie Kieffer
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Fahd Hilal
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Anne-Sophie Gay
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Delphine Debayle
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Marie Pronot
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Gwénola Poupon
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Iliona Lacagne
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, Institut National de la Santé Et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Stéphane Martin
- Université Côte d'Azur, Institut National de la Santé Et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Carole Gwizdek
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- *Correspondence: Carole Gwizdek,
| |
Collapse
|
7
|
Corrêa-Velloso JC, Linardi AM, Glaser T, Velloso FJ, Rivas MP, Leite REP, Grinberg LT, Ulrich H, Akins MR, Chiavegatto S, Haddad LA. Fmr1 exon 14 skipping in late embryonic development of the rat forebrain. BMC Neurosci 2022; 23:32. [PMID: 35641906 PMCID: PMC9158170 DOI: 10.1186/s12868-022-00711-1] [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: 12/06/2021] [Accepted: 04/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fragile X syndrome, the major cause of inherited intellectual disability among men, is due to deficiency of the synaptic functional regulator FMR1 protein (FMRP), encoded by the FMRP translational regulator 1 (FMR1) gene. FMR1 alternative splicing produces distinct transcripts that may consequently impact FMRP functional roles. In transcripts without exon 14 the translational reading frame is shifted. For deepening current knowledge of the differential expression of Fmr1 exon 14 along the rat nervous system development, we conducted a descriptive study employing quantitative RT-PCR and BLAST of RNA-Seq datasets. RESULTS We observed in the rat forebrain progressive decline of total Fmr1 mRNA from E11 to P112 albeit an elevation on P3; and exon-14 skipping in E17-E20 with downregulation of the resulting mRNA. We tested if the reduced detection of messages without exon 14 could be explained by nonsense-mediated mRNA decay (NMD) vulnerability, but knocking down UPF1, a major component of this pathway, did not increase their quantities. Conversely, it significantly decreased FMR1 mRNA having exon 13 joined with either exon 14 or exon 15 site A. CONCLUSIONS The forebrain in the third embryonic week of the rat development is a period with significant skipping of Fmr1 exon 14. This alternative splicing event chronologically precedes a reduction of total Fmr1 mRNA, suggesting that it may be part of combinatorial mechanisms downregulating the gene's expression in the late embryonic period. The decay of FMR1 mRNA without exon 14 should be mediated by a pathway different from NMD. Finally, we provide evidence of FMR1 mRNA stabilization by UPF1, likely depending on FMRP.
Collapse
Affiliation(s)
- Juliana C Corrêa-Velloso
- Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão, 277 # 327, São Paulo, SP, 05508-090, Brazil
| | - Alessandra M Linardi
- Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão, 277 # 327, São Paulo, SP, 05508-090, Brazil
| | - Talita Glaser
- Department of Biochemistry, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Fernando J Velloso
- Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão, 277 # 327, São Paulo, SP, 05508-090, Brazil
| | - Maria P Rivas
- Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão, 277 # 327, São Paulo, SP, 05508-090, Brazil
| | - Renata E P Leite
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Lea T Grinberg
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Michael R Akins
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Silvana Chiavegatto
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.,Department of Psychiatry, Instituto de Psiquiatria, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luciana A Haddad
- Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, Rua do Matão, 277 # 327, São Paulo, SP, 05508-090, Brazil.
| |
Collapse
|
8
|
Adayev T, LaFauci G, Xu W, Dobkin C, Kascsak R, Brown WT, Goodman JH. Development of a Quantitative FMRP Assay for Mouse Tissue Applications. Genes (Basel) 2021; 12:genes12101516. [PMID: 34680911 PMCID: PMC8535242 DOI: 10.3390/genes12101516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 11/20/2022] Open
Abstract
Fragile X syndrome results from the absence of the FMR1 gene product—Fragile X Mental Retardation Protein (FMRP). Fragile X animal research has lacked a reliable method to quantify FMRP. We report the development of an array of FMRP-specific monoclonal antibodies and their application for quantitative assessment of FMRP (qFMRPm) in mouse tissue. To characterize the assay, we determined the normal variability of FMRP expression in four brain structures of six different mouse strains at seven weeks of age. There was a hierarchy of FMRP expression: neocortex > hippocampus > cerebellum > brainstem. The expression of FMRP was highest and least variable in the neocortex, whereas it was most variable in the hippocampus. Male C57Bl/6J and FVB mice were selected to determine FMRP developmental differences in the brain at 3, 7, 10, and 14 weeks of age. We examined the four structures and found a developmental decline in FMRP expression with age, except for the brainstem where it remained stable. qFMRPm assay of blood had highest values in 3 week old animals and dropped by 2.5-fold with age. Sex differences were not significant. The results establish qFMRPm as a valuable tool due to its ease of methodology, cost effectiveness, and accuracy.
Collapse
Affiliation(s)
- Tatyana Adayev
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
- Correspondence: ; Tel.: +1-718-494-5314
| | - Giuseppe LaFauci
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - Weimin Xu
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - Carl Dobkin
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - Richard Kascsak
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - W. Ted Brown
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
- Perkins Center, University of Sydney Camperdown, Sydney, NSW 2006, Australia
| | - Jeffrey H. Goodman
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| |
Collapse
|
9
|
Hooper AW, Wong H, Niibori Y, Abdoli R, Karumuthil-Melethil S, Qiao C, Danos O, Bruder JT, Hampson DR. Gene therapy using an ortholog of human fragile X mental retardation protein partially rescues behavioral abnormalities and EEG activity. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:196-209. [PMID: 34485605 PMCID: PMC8399347 DOI: 10.1016/j.omtm.2021.06.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/30/2021] [Indexed: 01/28/2023]
Abstract
Fragile X syndrome (FXS), a neurodevelopmental disorder with no known cure, is caused by a lack of expression of the fragile X mental retardation protein (FMRP). As a single-gene disorder, FXS is an excellent candidate for viral-vector-based gene therapy, although that is complicated by the existence of multiple isoforms of FMRP, whose individual cellular functions are unknown. We studied the effects of rat and mouse orthologs of human isoform 17, a major expressed isoform of FMRP. Injection of neonatal Fmr1 knockout rats and mice with adeno-associated viral vectors (AAV9 serotype) under the control of an MeCP2 mini-promoter resulted in widespread distribution of the FMRP transgenes throughout the telencephalon and diencephalon. Transgene expression occurred mainly in non-GABAergic neurons, with little expression in glia. Early postnatal treatment resulted in partial rescue of the Fmr1 KO rat phenotype, including improved social dominance in treated Fmr1 KO females and partial rescue of locomotor activity in males. Electro-encephalogram (EEG) recordings showed correction of abnormal slow-wave activity during the sleep-like state in male Fmr1 KO rats. These findings support the use of AAV-based gene therapy as a treatment for FXS and specifically demonstrate the potential therapeutic benefit of human FMRP isoform 17 orthologs.
Collapse
Affiliation(s)
- Alexander W.M. Hooper
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | - Hayes Wong
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | - Yosuke Niibori
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | - Rozita Abdoli
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada M5S 3M2
| | | | - Chunping Qiao
- Research and Early Development, REGENXBIO Inc. Rockville, Maryland, U.S.A. 20850
| | - Olivier Danos
- Research and Early Development, REGENXBIO Inc. Rockville, Maryland, U.S.A. 20850
| | - Joseph T. Bruder
- Research and Early Development, REGENXBIO Inc. Rockville, Maryland, U.S.A. 20850
| | - David R. Hampson
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada M5S 3M2
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 3M2
- Corresponding author: David R. Hampson, PhD, Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, Univerity of Toronto, ON M5S 3M2, Canada.
| |
Collapse
|
10
|
Dionne O, Corbin F. An "Omic" Overview of Fragile X Syndrome. BIOLOGY 2021; 10:433. [PMID: 34068266 PMCID: PMC8153138 DOI: 10.3390/biology10050433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/01/2021] [Accepted: 05/08/2021] [Indexed: 01/16/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder associated with a wide range of cognitive, behavioral and medical problems. It arises from the silencing of the fragile X mental retardation 1 (FMR1) gene and, consequently, in the absence of its encoded protein, FMRP (fragile X mental retardation protein). FMRP is a ubiquitously expressed and multifunctional RNA-binding protein, primarily considered as a translational regulator. Pre-clinical studies of the past two decades have therefore focused on this function to relate FMRP's absence to the molecular mechanisms underlying FXS physiopathology. Based on these data, successful pharmacological strategies were developed to rescue fragile X phenotype in animal models. Unfortunately, these results did not translate into humans as clinical trials using same therapeutic approaches did not reach the expected outcomes. These failures highlight the need to put into perspective the different functions of FMRP in order to get a more comprehensive understanding of FXS pathophysiology. This work presents a review of FMRP's involvement on noteworthy molecular mechanisms that may ultimately contribute to various biochemical alterations composing the fragile X phenotype.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, QC J1H 5H4, Canada;
| | | |
Collapse
|
11
|
Titus MB, Wright EG, Bono JM, Poliakon AK, Goldstein BR, Super MK, Young LA, Manaj M, Litchford M, Reist NE, Killian DJ, Olesnicky EC. The conserved alternative splicing factor caper regulates neuromuscular phenotypes during development and aging. Dev Biol 2021; 473:15-32. [PMID: 33508255 PMCID: PMC7987824 DOI: 10.1016/j.ydbio.2021.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/17/2022]
Abstract
RNA-binding proteins play an important role in the regulation of post-transcriptional gene expression throughout the nervous system. This is underscored by the prevalence of mutations in genes encoding RNA splicing factors and other RNA-binding proteins in a number of neurodegenerative and neurodevelopmental disorders. The highly conserved alternative splicing factor Caper is widely expressed throughout the developing embryo and functions in the development of various sensory neural subtypes in the Drosophila peripheral nervous system. Here we find that caper dysfunction leads to aberrant neuromuscular junction morphogenesis, as well as aberrant locomotor behavior during larval and adult stages. Despite its widespread expression, our results indicate that caper function is required to a greater extent within the nervous system, as opposed to muscle, for neuromuscular junction development and for the regulation of adult locomotor behavior. Moreover, we find that Caper interacts with the RNA-binding protein Fmrp to regulate adult locomotor behavior. Finally, we show that caper dysfunction leads to various phenotypes that have both a sex and age bias, both of which are commonly seen in neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- M Brandon Titus
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Ethan G Wright
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Jeremy M Bono
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Andrea K Poliakon
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Brandon R Goldstein
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Meg K Super
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Lauren A Young
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Melpomeni Manaj
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Morgan Litchford
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Noreen E Reist
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Darrell J Killian
- Department of Molecular Biology, Colorado College, Colorado Springs, CO, 80903, USA
| | - Eugenia C Olesnicky
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA.
| |
Collapse
|
12
|
Malecki C, Hambly BD, Jeremy RW, Robertson EN. The RNA-binding fragile-X mental retardation protein and its role beyond the brain. Biophys Rev 2020; 12:903-916. [PMID: 32654068 DOI: 10.1007/s12551-020-00730-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
It is well-established that variations of a CGG repeat expansion in the gene FMR1, which encodes the fragile-X mental retardation protein (FMRP), cause the neurocognitive disorder, fragile-X syndrome (FXS). However, multiple observations suggest a general and complex regulatory role of FMRP in processes outside the brain: (1) FMRP is ubiquitously expressed in the body, suggesting it functions in multiple organ systems; (2) patients with FXS can exhibit a physical phenotype that is consistent with an underlying abnormality in connective tissue; (3) different CGG repeat expansion lengths in FMR1 result in different clinical outcomes due to different pathogenic mechanisms; (4) the function of FMRP as an RNA-binding protein suggests it has a general regulatory role. This review details the complex nature of FMRP and the different CGG repeat expansion lengths and the evidence supporting the essential role of the protein in a variety of biological and pathological processes.
Collapse
Affiliation(s)
- Cassandra Malecki
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia.
| | - Brett D Hambly
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia
| | - Richmond W Jeremy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia.,Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Elizabeth N Robertson
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia.,Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
13
|
Zafarullah M, Tang HT, Durbin-Johnson B, Fourie E, Hessl D, Rivera SM, Tassone F. FMR1 locus isoforms: potential biomarker candidates in fragile X-associated tremor/ataxia syndrome (FXTAS). Sci Rep 2020; 10:11099. [PMID: 32632326 PMCID: PMC7338407 DOI: 10.1038/s41598-020-67946-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Fragile X associated tremor/ataxia syndrome (FXTAS) is a late adult-onset neurodegenerative disorder that affects movement and cognition in male and female carriers of a premutation allele of 55-200 CGG repeats in the Fragile X mental retardation (FMR1) gene. It is currently unknown if and when an individual carrier of a premutation allele will develop FXTAS, as clinical assessment fails to identify carriers at risk before significant neurological symptoms are evident. The primary objective of this study was to investigate the alternative splicing landscape at the FMR1 locus in conjunction with brain measures in male individuals with a premutation allele enrolled in a very first longitudinal study, compared to age-matched healthy male controls, with the purpose of identifying biomarkers for early diagnosis, disease prediction and, a progression of FXTAS. Our findings indicate that increased expression of FMR1 mRNA isoforms, including Iso4/4b, Iso10/10b, as well as of the ASFMR1 mRNAs Iso131bp, are present in premutation carriers as compared to non-carrier healthy controls. More specifically, we observed a higher expression of Iso4/4b and Iso10/10b, which encode for truncated proteins, only in those premutation carriers who developed symptoms of FXTAS over time as compared to non-carrier healthy controls, suggesting a potential role in the development of the disorder. In addition, we found a significant association of these molecular changes with various measurements of brain morphology, including the middle cerebellar peduncle (MCP), superior cerebellar peduncle (SCP), pons, and midbrain, indicating their potential contribution to the pathogenesis of FXTAS. Interestingly, the high expression levels of Iso4/4b observed both at visit 1 and visit 2 and found to be associated with a decrease in mean MCP width only in those individuals who developed FXTAS over time, suggests their role as potential biomarkers for early diagnosis of FXTAS.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817 CA, USA
| | - Hiu-Tung Tang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817 CA, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, School of Medicine, University of California Davis, Davis, CA, USA
| | - Emily Fourie
- Center for Mind and Brain, University of California Davis, Davis, CA, USA
- Department of Psychology, University of California, Davis, Davis, CA, USA
| | - David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, 95817 CA, USA
| | - Susan M Rivera
- Center for Mind and Brain, University of California Davis, Davis, CA, USA
- Department of Psychology, University of California, Davis, Davis, CA, USA
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, 95817 CA, USA.
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA.
| |
Collapse
|
14
|
Yang WJ, Yan AZ, Xu YJ, Guo XY, Fu XG, Li D, Liao J, Zhang D, Lan FH. Further identification of a 140bp sequence from amid intron 9 of human FMR1 gene as a new exon. BMC Genet 2020; 21:63. [PMID: 32552710 PMCID: PMC7301526 DOI: 10.1186/s12863-020-00870-2] [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: 01/07/2020] [Accepted: 06/09/2020] [Indexed: 11/24/2022] Open
Abstract
Background The disease gene of fragile X syndrome, FMR1 gene, encodes fragile X mental retardation protein (FMRP). The alternative splicing (AS) of FMR1 can affect the structure and function of FMRP. However, the biological functions of alternatively spliced isoforms remain elusive. In a previous study, we identified a new 140bp exon from the intron 9 of human FMR1 gene. In this study, we further examined the biological functions of this new exon and its underlying signaling pathways. Results qRT-PCR results showed that this novel exon is commonly expressed in the peripheral blood of normal individuals. Comparative genomics showed that sequences paralogous to the 140 bp sequence only exist in the genomes of primates. To explore the biological functions of the new transcript, we constructed recombinant eukaryotic expression vectors and lentiviral overexpression vectors. Results showed that the spliced transcript encoded a truncated protein which was expressed mainly in the cell nucleus. Additionally, several genes, including the BEX1 gene involved in mGluR-LTP or mGluR-LTD signaling pathways were significantly influenced when the truncated FMRP was overexpressed. Conclusions our work identified a new exon from amid intron 9 of human FMR1 gene with wide expression in normal healthy individuals, which emphasizes the notion that the AS of FMR1 gene is complex and may in a large part account for the multiple functions of FMRP.
Collapse
Affiliation(s)
- Wen-Jing Yang
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China.,Present addresses: Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ai-Zhen Yan
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China
| | - Yong-Jun Xu
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China
| | - Xiao-Yan Guo
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China.,Present addresses: Department of Laboratory Medicine, Fuzhou No. 2 Hospital Affiliated Xiamen University, Fuzhou, Fujian, 350007, People's Republic of China
| | - Xian-Guo Fu
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China.,Present addresses: Department of Laboratory Medicine, Ningde Municipal Hospital, Fujian Medical University, Ningde City, 352100, Fujian Province, China
| | - Dan Li
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China
| | - Juan Liao
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China.,Present addresses: Department of Laboratory Medicine, Fujian University of Traditional Chinese Medicine Affiliated People's Hospital, Fuzhou, 350001, Fujian, China
| | - Duo Zhang
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China
| | - Feng-Hua Lan
- Department of Clinical Genetics and Experimental Medicine, 900th Hospital of the Joint Logistics Force, Xiamen University School of Medicine, 156 Xi'erhuanbei Road, Fuzhou City, Fujian Province, 350025, People's Republic of China.
| |
Collapse
|
15
|
Suardi GAM, Haddad LA. FMRP ribonucleoprotein complexes and RNA homeostasis. ADVANCES IN GENETICS 2020; 105:95-136. [PMID: 32560791 DOI: 10.1016/bs.adgen.2020.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Fragile Mental Retardation 1 gene (FMR1), at Xq27.3, encodes the fragile mental retardation protein (FMRP), and displays in its 5'-untranslated region a series of polymorphic CGG triplet repeats that may undergo dynamic mutation. Fragile X syndrome (FXS) is the leading cause of inherited intellectual disability among men, and is most frequently due to FMR1 full mutation and consequent transcription repression. FMR1 premutations may associate with at least two other clinical conditions, named fragile X-associated primary ovarian insufficiency (FXPOI) and tremor and ataxia syndrome (FXTAS). While FXPOI and FXTAS appear to be mediated by FMR1 mRNA accumulation, relative reduction of FMRP, and triplet repeat translation, FXS is due to the lack of the RNA-binding protein FMRP. Besides its function as mRNA translation repressor in neuronal and stem/progenitor cells, RNA editing roles have been assigned to FMRP. In this review, we provide a brief description of FMR1 transcribed microsatellite and associated clinical disorders, and discuss FMRP molecular roles in ribonucleoprotein complex assembly and trafficking, as well as aspects of RNA homeostasis affected in FXS cells.
Collapse
Affiliation(s)
- Gabriela Aparecida Marcondes Suardi
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Luciana Amaral Haddad
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
16
|
Sharon G, Cruz NJ, Kang DW, Gandal MJ, Wang B, Kim YM, Zink EM, Casey CP, Taylor BC, Lane CJ, Bramer LM, Isern NG, Hoyt DW, Noecker C, Sweredoski MJ, Moradian A, Borenstein E, Jansson JK, Knight R, Metz TO, Lois C, Geschwind DH, Krajmalnik-Brown R, Mazmanian SK. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019; 177:1600-1618.e17. [PMID: 31150625 PMCID: PMC6993574 DOI: 10.1016/j.cell.2019.05.004] [Citation(s) in RCA: 640] [Impact Index Per Article: 128.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/11/2019] [Accepted: 04/30/2019] [Indexed: 01/02/2023]
Abstract
Autism spectrum disorder (ASD) manifests as alterations in complex human behaviors including social communication and stereotypies. In addition to genetic risks, the gut microbiome differs between typically developing (TD) and ASD individuals, though it remains unclear whether the microbiome contributes to symptoms. We transplanted gut microbiota from human donors with ASD or TD controls into germ-free mice and reveal that colonization with ASD microbiota is sufficient to induce hallmark autistic behaviors. The brains of mice colonized with ASD microbiota display alternative splicing of ASD-relevant genes. Microbiome and metabolome profiles of mice harboring human microbiota predict that specific bacterial taxa and their metabolites modulate ASD behaviors. Indeed, treatment of an ASD mouse model with candidate microbial metabolites improves behavioral abnormalities and modulates neuronal excitability in the brain. We propose that the gut microbiota regulates behaviors in mice via production of neuroactive metabolites, suggesting that gut-brain connections contribute to the pathophysiology of ASD.
Collapse
Affiliation(s)
- Gil Sharon
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Nikki Jamie Cruz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Dae-Wook Kang
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ 85287, USA; Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA
| | - Michael J Gandal
- Center for Autism Research and Treatment, Program in Neurobehavioral Genetics, Semel Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, Semel Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Young-Mo Kim
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Erika M Zink
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Cameron P Casey
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Bryn C Taylor
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christianne J Lane
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Lisa M Bramer
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Nancy G Isern
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - David W Hoyt
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Cecilia Noecker
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Michael J Sweredoski
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Annie Moradian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Elhanan Borenstein
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Department of Computer Science and Engineering, University of Washington, Seattle, WA 98195, USA; Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 6997801, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Janet K Jansson
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biongineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thomas O Metz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Daniel H Geschwind
- Center for Autism Research and Treatment, Program in Neurobehavioral Genetics, Semel Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, Semel Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ 85287, USA; Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85287, USA; School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ 85287, USA
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
17
|
The Application of Adeno-Associated Viral Vector Gene Therapy to the Treatment of Fragile X Syndrome. Brain Sci 2019; 9:brainsci9020032. [PMID: 30717399 PMCID: PMC6406794 DOI: 10.3390/brainsci9020032] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/18/2023] Open
Abstract
Viral vector-mediated gene therapy has grown by leaps and bounds over the past several years. Although the reasons for this progress are varied, a deeper understanding of the basic biology of the viruses, the identification of new and improved versions of viral vectors, and simply the vast experience gained by extensive testing in both animal models of disease and in clinical trials, have been key factors. Several studies have investigated the efficacy of adeno-associated viral (AAV) vectors in the mouse model of fragile X syndrome where AAVs have been used to express fragile X mental retardation protein (FMRP), which is missing or highly reduced in the disorder. These studies have demonstrated a range of efficacies in different tests from full correction, to partial rescue, to no effect. Here we provide a backdrop of recent advances in AAV gene therapy as applied to central nervous system disorders, outline the salient features of the fragile X studies, and discuss several key issues for moving forward. Collectively, the findings to date from the mouse studies on fragile X syndrome, and data from clinical trials testing AAVs in other neurological conditions, indicate that AAV-mediated gene therapy could be a viable strategy for treating fragile X syndrome.
Collapse
|
18
|
Wang Q, Barad DH, Darmon SK, Kushnir VA, Wu YG, Lazzaroni-Tealdi E, Zhang L, Albertini DF, Gleicher N. Reduced RNA expression of the FMR1 gene in women with low (CGGn<26) repeats. PLoS One 2018; 13:e0209309. [PMID: 30576349 PMCID: PMC6303073 DOI: 10.1371/journal.pone.0209309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/04/2018] [Indexed: 11/18/2022] Open
Abstract
Low FMR1 variants (CGGn<26) have been associated with premature ovarian aging, female infertility and poor IVF treatment success. Until now, there is little published information concerning possible molecular mechanisms for this effect. We wished to examine whether relative expression of RNA and the FMR1 gene’s fragile X mental retardation protein (FMRP) RNA isoforms differ in women with various FMR1 sub-genotypes (normal, low CGGn<26 and/or high CGGn≥34). This prospective cohort study was conducted between 2014 and 2017 in a clinical research unit of the Center for Human Reproduction in New York City. The study involved a total of 98 study subjects, including 18 young oocyte donors and 80 older infertility patients undergoing routine in vitro fertilization (IVF) cycles. The main outcome measure was RNA expression in human luteinized granulosa cells of 5 groups of FMRP isoforms. The relative expression of FMR1 RNA in human luteinized granulosa cells was measured by real-time PCR and a possible association with CGGn was explored. All 5 groups of FMRP RNA isoforms examined were found to be differentially expressed in human luteinized granulosa cells. The relative expression of four FMR1 RNA isoforms showed significant differences among 6 FMR1 sub-genotypes. Women with at least one low allele expressed significantly lower levels of all 5 sets of FRMP isoforms in comparison to the non-low group. While it would be of interest to see whether FMRP is also decreased in the low-group we recognize that in recent years it has been increasingly documented that information flow of genetics may be regulated by non-coding RNA, that is, without translation to a protein product. We, thus, conclude that various CGG expansions of FMR1 allele may lead to changes of RNA levels and ratios of distinct FMRP RNA isoforms, which could regulate the translation and/or cellular localization of FMRP, affect the expression of steroidogenic enzymes and hormonal receptors, or act in some other epigenetic process and therefore result in the ovarian dysfunction in infertility.
Collapse
Affiliation(s)
- Qi Wang
- The Center for Human Reproduction, New York, NY, United States of America
| | - David H. Barad
- The Center for Human Reproduction, New York, NY, United States of America
- The Foundation for Reproductive Medicine, New York, NY, United States of America
- * E-mail:
| | - Sarah K. Darmon
- The Center for Human Reproduction, New York, NY, United States of America
| | - Vitaly A. Kushnir
- The Center for Human Reproduction, New York, NY, United States of America
- Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, NC, United States of America
| | - Yan-Guang Wu
- The Center for Human Reproduction, New York, NY, United States of America
| | | | - Lin Zhang
- The Center for Human Reproduction, New York, NY, United States of America
| | - David F. Albertini
- The Center for Human Reproduction, New York, NY, United States of America
- Department of Molecular and Integrative Physiology, University of Kansas Hospital, Kansas City, KS, United States of America
| | - Norbert Gleicher
- The Center for Human Reproduction, New York, NY, United States of America
- The Foundation for Reproductive Medicine, New York, NY, United States of America
- Stem Cell and Molecular Embryology Laboratory, the Rockefeller University, New York, NY, United States of America
- Department of Obstetrics and Gynecology, Vienna University School of Medicine, Vienna, Austria
| |
Collapse
|
19
|
Dockendorff TC, Labrador M. The Fragile X Protein and Genome Function. Mol Neurobiol 2018; 56:711-721. [PMID: 29796988 DOI: 10.1007/s12035-018-1122-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022]
Abstract
The fragile X syndrome (FXS) arises from loss of expression or function of the FMR1 gene and is one of the most common monogenic forms of intellectual disability and autism. During the past two decades of FXS research, the fragile X mental retardation protein (FMRP) has been primarily characterized as a cytoplasmic RNA binding protein that facilitates transport of select RNA substrates through neural projections and regulation of translation within synaptic compartments, with the protein products of such mRNAs then modulating cognitive functions. However, the presence of a small fraction of FMRP in the nucleus has long been recognized. Accordingly, recent studies have uncovered several mechanisms or pathways by which FMRP influences nuclear gene expression and genome function. Some of these pathways appear to be independent of the classical role for FMRP as a regulator of translation and point to novel functions, including the possibility that FMRP directly participates in the DNA damage response and in the maintenance of genome stability. In this review, we highlight these advances and discuss how these new findings could contribute to our understanding of FMRP in brain development and function, the neural pathology of fragile X syndrome, and perhaps impact of future therapeutic considerations.
Collapse
Affiliation(s)
- Thomas C Dockendorff
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA.
| | - Mariano Labrador
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
20
|
Khayachi A, Gwizdek C, Poupon G, Alcor D, Chafai M, Cassé F, Maurin T, Prieto M, Folci A, De Graeve F, Castagnola S, Gautier R, Schorova L, Loriol C, Pronot M, Besse F, Brau F, Deval E, Bardoni B, Martin S. Sumoylation regulates FMRP-mediated dendritic spine elimination and maturation. Nat Commun 2018; 9:757. [PMID: 29472612 PMCID: PMC5823917 DOI: 10.1038/s41467-018-03222-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/28/2018] [Indexed: 12/02/2022] Open
Abstract
Fragile X syndrome (FXS) is the most frequent inherited cause of intellectual disability and the best-studied monogenic cause of autism. FXS results from the functional absence of the fragile X mental retardation protein (FMRP) leading to abnormal pruning and consequently to synaptic communication defects. Here we show that FMRP is a substrate of the small ubiquitin-like modifier (SUMO) pathway in the brain and identify its active SUMO sites. We unravel the functional consequences of FMRP sumoylation in neurons by combining molecular replacement strategy, biochemical reconstitution assays with advanced live-cell imaging. We first demonstrate that FMRP sumoylation is promoted by activation of metabotropic glutamate receptors. We then show that this increase in sumoylation controls the homomerization of FMRP within dendritic mRNA granules which, in turn, regulates spine elimination and maturation. Altogether, our findings reveal the sumoylation of FMRP as a critical activity-dependent regulatory mechanism of FMRP-mediated neuronal function. Fragile X syndrome patients display intellectual disability and autism, caused by mutations in the RNA-binding protein fragile X mental retardation protein (FMRP). Here, the authors show that FMRP sumoylation is required for regulating spine density and maturation.
Collapse
Affiliation(s)
| | - Carole Gwizdek
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Gwénola Poupon
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Damien Alcor
- Université Côte d'Azur, INSERM, C3M, 06200, Nice, France
| | - Magda Chafai
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Frédéric Cassé
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Thomas Maurin
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Marta Prieto
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | | | | | | | - Romain Gautier
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Lenka Schorova
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Céline Loriol
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Marie Pronot
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Florence Besse
- Université Côte d'Azur, CNRS, INSERM, iBV, 06108, Nice, France
| | - Frédéric Brau
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Emmanuel Deval
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, INSERM, CNRS, IPMC, 06560, Valbonne, France
| | - Stéphane Martin
- Université Côte d'Azur, INSERM, CNRS, IPMC, 06560, Valbonne, France.
| |
Collapse
|
21
|
Vlassakis J, Herr AE. Joule Heating-Induced Dispersion in Open Microfluidic Electrophoretic Cytometry. Anal Chem 2017; 89:12787-12796. [PMID: 29110464 DOI: 10.1021/acs.analchem.7b03096] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
While protein electrophoresis conducted in capillaries and microchannels offers high-resolution separations, such formats can be cumbersome to parallelize for single-cell analysis. One approach for realizing large numbers of concurrent separations is open microfluidics (i.e., no microchannels). In an open microfluidic device adapted for single-cell electrophoresis, we perform 100s to 1000s of simultaneous separations of endogenous proteins. The microscope slide-sized device contains cells isolated in microwells located in a ∼40 μm polyacrylamide gel. The gel supports protein electrophoresis after concurrent in situ chemical lysis of each isolated cell. During electrophoresis, Joule (or resistive) heating degrades separation performance. Joule heating effects are expected to be acute in open microfluidic devices, where a single, high-conductivity buffer expedites the transition from cell lysis to protein electrophoresis. Here, we test three key assertions. First, Joule heating substantially impacts analytical sensitivity due to diffusive losses of protein out of the open microfluidic electrophoretic (EP) cytometry device. Second, increased analyte diffusivity due to autothermal runaway Joule heating is a dominant mechanism that reduces separation resolution in EP cytometry. Finally, buffer exchange reduces diffusive losses and band broadening, even when handling single-cell lysate protein concentrations in an open device. We develop numerical simulations of Joule heating-enhanced diffusion during electrophoresis and observe ∼50% protein loss out of the gel, which is reduced using the buffer exchange. Informed by analytical model predictions of separation resolution (with Joule heating), we empirically demonstrate nearly fully resolved separations of proteins with molecular mass differences of just 4 kDa or 12% (GAPDH, 36 kDa; PS6, 32 kDa) in each of 129 single cells. The attained separation performance with buffer exchange is relevant to detection of currently unmeasurable protein isoforms responsible for cancer progression.
Collapse
Affiliation(s)
- Julea Vlassakis
- Department of Bioengineering and ‡The UC Berkeley/UCSF Graduate Program in Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| | - Amy E Herr
- Department of Bioengineering and ‡The UC Berkeley/UCSF Graduate Program in Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| |
Collapse
|
22
|
Tseng E, Tang HT, AlOlaby RR, Hickey L, Tassone F. Altered expression of the FMR1 splicing variants landscape in premutation carriers. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:1117-1126. [PMID: 28888471 DOI: 10.1016/j.bbagrm.2017.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/26/2017] [Accepted: 08/26/2017] [Indexed: 01/17/2023]
Abstract
FMR1 premutation carriers (55-200 CGG repeats) are at risk for developing Fragile X-associated Tremor/Ataxia Syndrome (FXTAS), an adult onset neurodegenerative disorder. Approximately 20% of female carriers will develop Fragile X-associated Primary Ovarian Insufficiency (FXPOI), in addition to a number of clinical problems affecting premutation carriers throughout their life span. Marked elevation in FMR1 mRNA levels have been observed with premutation alleles resulting in RNA toxicity, the leading molecular mechanism proposed for the FMR1 associated disorders observed in premutation carriers. The FMR1 gene undergoes alternative splicing and we have recently reported that the relative abundance of all FMR1 mRNA isoforms is significantly increased in premutation carriers. In this study, we characterized the transcriptional FMR1 isoforms distribution pattern in different tissues and identified a total of 49 isoforms, some of which observed only in premutation carriers and which might play a role in the pathogenesis of FXTAS. Further, we investigated the distribution pattern and expression levels of the FMR1 isoforms in asymptomatic premutation carriers and in those with FXTAS and found no significant differences between the two groups. Our findings suggest that the characterization of the expression levels of the different FMR1 isoforms is fundamental for understanding the regulation of the FMR1 gene as imbalance in their expression could lead to an altered functional diversity with neurotoxic consequences. Their characterization will also help to elucidating the mechanism(s) by which "toxic gain of function" of the FMR1 mRNA may play a role in FXTAS and/or in the other FMR1-associated conditions.
Collapse
Affiliation(s)
| | - Hiu-Tung Tang
- Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Reem Rafik AlOlaby
- Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Luke Hickey
- Pacific Biosciences, Inc., Menlo Park, CA 94025, USA
| | - Flora Tassone
- Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, USA; MIND Institute, UC Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
23
|
McAninch DS, Heinaman AM, Lang CN, Moss KR, Bassell GJ, Rita Mihailescu M, Evans TL. Fragile X mental retardation protein recognizes a G quadruplex structure within the survival motor neuron domain containing 1 mRNA 5'-UTR. MOLECULAR BIOSYSTEMS 2017; 13:1448-1457. [PMID: 28612854 PMCID: PMC5544254 DOI: 10.1039/c7mb00070g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
G quadruplex structures have been predicted by bioinformatics to form in the 5'- and 3'-untranslated regions (UTRs) of several thousand mature mRNAs and are believed to play a role in translation regulation. Elucidation of these roles has primarily been focused on the 3'-UTR, with limited focus on characterizing the G quadruplex structures and functions in the 5'-UTR. Investigation of the affinity and specificity of RNA binding proteins for 5'-UTR G quadruplexes and the resulting regulatory effects have also been limited. Among the mRNAs predicted to form a G quadruplex structure within the 5'-UTR is the survival motor neuron domain containing 1 (SMNDC1) mRNA, encoding a protein that is critical to the spliceosome. Additionally, this mRNA has been identified as a potential target of the fragile X mental retardation protein (FMRP), whose loss of expression leads to fragile X syndrome. FMRP is an RNA binding protein involved in translation regulation that has been shown to bind mRNA targets that form G quadruplex structures. In this study we have used biophysical methods to investigate G quadruplex formation in the 5'-UTR of SMNDC1 mRNA and analyzed its interactions with FMRP. Our results show that SMNDC1 mRNA 5'-UTR forms an intramolecular, parallel G quadruplex structure comprised of three G quartet planes, which is bound specifically by FMRP both in vitro and in mouse brain lysates. These findings suggest a model by which FMRP might regulate the translation of a subset of its mRNA targets by recognizing the G quadruplex structure present in their 5'-UTR, and affecting their accessibility by the protein synthesis machinery.
Collapse
Affiliation(s)
- Damian S McAninch
- Department of Chemistry and Biochemistry, Duquesne University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, USA.
| | - Ashley M Heinaman
- Department of Chemistry, University of Pittsburgh at Johnstown, Johnstown, Pennsylvania 15904, USA
| | - Cara N Lang
- Department of Chemistry, University of Pittsburgh at Johnstown, Johnstown, Pennsylvania 15904, USA
| | - Kathryn R Moss
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Mihaela Rita Mihailescu
- Department of Chemistry and Biochemistry, Duquesne University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, USA.
| | - Timothy L Evans
- Department of Chemistry and Biochemistry, Duquesne University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, USA. and Department of Chemistry, University of Pittsburgh at Johnstown, Johnstown, Pennsylvania 15904, USA
| |
Collapse
|
24
|
Lu P, Chen X, Feng Y, Zeng Q, Jiang C, Zhu X, Fan G, Xue Z. Integrated transcriptome analysis of human iPS cells derived from a fragile X syndrome patient during neuronal differentiation. SCIENCE CHINA. LIFE SCIENCES 2016; 59:1093-1105. [PMID: 27730449 DOI: 10.1007/s11427-016-0194-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/05/2016] [Indexed: 01/01/2023]
Abstract
Fragile X syndrome (FXS) patients carry the expansion of over 200 CGG repeats at the promoter of fragile X mental retardation 1 (FMR1), leading to decreased or absent expression of its encoded fragile X mental retardation protein (FMRP). However, the global transcriptional alteration by FMRP deficiency has not been well characterized at single nucleotide resolution, i.e., RNA-seq. Here, we performed in-vitro neuronal differentiation of human induced pluripotent stem (iPS) cells that were derived from fibroblasts of a FXS patient (FXS-iPSC). We then performed RNA-seq and examined the transcriptional misregulation at each intermediate stage during in-vitro differentiation of FXS-iPSC into neurons. After thoroughly analyzing the transcriptomic data and integrating them with those from other platforms, we found up-regulation of many genes encoding TFs for neuronal differentiation (WNT1, BMP4, POU3F4, TFAP2C, and PAX3), down-regulation of potassium channels (KCNA1, KCNC3, KCNG2, KCNIP4, KCNJ3, KCNK9, and KCNT1) and altered temporal regulation of SHANK1 and NNAT in FXS-iPSC derived neurons, indicating impaired neuronal differentiation and function in FXS patients. In conclusion, we demonstrated that the FMRP deficiency in FXS patients has significant impact on the gene expression patterns during development, which will help to discover potential targeting candidates for the cure of FXS symptoms.
Collapse
Affiliation(s)
- Ping Lu
- Tongji Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xiaolong Chen
- Tongji University, School of Life Sciences and Technology, Shanghai, 200092, China
| | - Yun Feng
- Tongji Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiao Zeng
- Tongji Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Cizhong Jiang
- Tongji University, School of Life Sciences and Technology, Shanghai, 200092, China
| | - Xianmin Zhu
- Tongji University, School of Life Sciences and Technology, Shanghai, 200092, China.
| | - Guoping Fan
- Tongji University, School of Life Sciences and Technology, Shanghai, 200092, China.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Zhigang Xue
- Tongji Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200065, China.
- Tongji University Suzhou Institute, Suzhou, 215101, China.
| |
Collapse
|
25
|
Zimmer SE, Doll SG, Garcia ADR, Akins MR. Splice form-dependent regulation of axonal arbor complexity by FMRP. Dev Neurobiol 2016; 77:738-752. [PMID: 27643955 DOI: 10.1002/dneu.22453] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/31/2016] [Accepted: 09/14/2016] [Indexed: 01/01/2023]
Abstract
The autism-related protein Fragile X mental retardation protein (FMRP) is an RNA binding protein that plays important roles during both nervous system development and experience dependent plasticity. Alternative splicing of the Fmr1 locus gives rise to 12 different FMRP splice forms that differ in the functional and regulatory domains they contain as well as in their expression profile among brain regions and across development. Complete loss of FMRP leads to morphological and functional changes in neurons, including an increase in the size and complexity of the axonal arbor. To investigate the relative contribution of the FMRP splice forms to the regulation of axon morphology, we overexpressed individual splice forms in cultured wild type rat cortical neurons. FMRP overexpression led to a decrease in axonal arbor complexity that suggests that FMRP regulates axon branching. This reduction in complexity was specific to three splice forms-the full-length splice form 1, the most highly expressed splice form 7, and splice form 9. A focused analysis of splice form 7 revealed that this regulation is independent of RNA binding. Instead this regulation is disrupted by mutations affecting phosphorylation of a conserved serine as well as by mutating the nuclear export sequence. Surprisingly, this mutation in the nuclear export sequence also led to increased localization to the distal axonal arbor. Together, these findings reveal domain-specific functions of FMRP in the regulation of axonal complexity that may be controlled by differential expression of FMRP splice forms. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 738-752, 2017.
Collapse
Affiliation(s)
| | - Steven G Doll
- Department of Biology, Drexel University, Philadelphia, Pennsylvania
| | - A Denise R Garcia
- Department of Biology, Drexel University, Philadelphia, Pennsylvania.,Department of Neurobiology and Anatomy, Drexel University, Philadelphia, Pennsylvania
| | - Michael R Akins
- Department of Biology, Drexel University, Philadelphia, Pennsylvania.,Department of Neurobiology and Anatomy, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Abbey M. Functional characterization of the several splice variants of Fmr1. RESEARCH IDEAS AND OUTCOMES 2016. [DOI: 10.3897/rio.2.e10593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
27
|
RNA Secondary Structure Modulates FMRP's Bi-Functional Role in the MicroRNA Pathway. Int J Mol Sci 2016; 17:ijms17060985. [PMID: 27338369 PMCID: PMC4926514 DOI: 10.3390/ijms17060985] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/23/2016] [Accepted: 06/14/2016] [Indexed: 11/28/2022] Open
Abstract
MicroRNAs act by post-transcriptionally regulating the gene expression of 30%–60% of mammalian genomes. MicroRNAs are key regulators in all cellular processes, though the mechanism by which the cell activates or represses microRNA-mediated translational regulation is poorly understood. In this review, we discuss the RNA binding protein Fragile X Mental Retardation Protein (FMRP) and its role in microRNA-mediated translational regulation. Historically, FMRP is known to function as a translational suppressor. However, emerging data suggests that FMRP has both an agonistic and antagonistic role in regulating microRNA-mediated translational suppression. This bi-functional role is dependent on FMRP’s interaction with the RNA helicase Moloney leukemia virus 10 (MOV10), which modifies the structural landscape of bound mRNA, therefore facilitating or inhibiting its association with the RNA-Induced Silencing Complex.
Collapse
|
28
|
Järvelin AI, Noerenberg M, Davis I, Castello A. The new (dis)order in RNA regulation. Cell Commun Signal 2016; 14:9. [PMID: 27048167 PMCID: PMC4822317 DOI: 10.1186/s12964-016-0132-3] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/21/2016] [Indexed: 02/03/2023] Open
Abstract
RNA-binding proteins play a key role in the regulation of all aspects of RNA metabolism, from the synthesis of RNA to its decay. Protein-RNA interactions have been thought to be mostly mediated by canonical RNA-binding domains that form stable secondary and tertiary structures. However, a number of pioneering studies over the past decades, together with recent proteome-wide data, have challenged this view, revealing surprising roles for intrinsically disordered protein regions in RNA binding. Here, we discuss how disordered protein regions can mediate protein-RNA interactions, conceptually grouping these regions into RS-rich, RG-rich, and other basic sequences, that can mediate both specific and non-specific interactions with RNA. Disordered regions can also influence RNA metabolism through protein aggregation and hydrogel formation. Importantly, protein-RNA interactions mediated by disordered regions can influence nearly all aspects of co- and post-transcriptional RNA processes and, consequently, their disruption can cause disease. Despite growing interest in disordered protein regions and their roles in RNA biology, their mechanisms of binding, regulation, and physiological consequences remain poorly understood. In the coming years, the study of these unorthodox interactions will yield important insights into RNA regulation in cellular homeostasis and disease.
Collapse
Affiliation(s)
- Aino I. Järvelin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU UK
| | - Marko Noerenberg
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU UK
| | - Ilan Davis
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU UK
| | - Alfredo Castello
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU UK
| |
Collapse
|
29
|
FU XIANGUO, ZHENG DEZHU, LIAO JUAN, LI QINGQIN, LIN YUXIANG, ZHANG DUO, YAN AIZHEN, LAN FENGHUA. Alternatively spliced products lacking exon 12 dominate the expression of fragile X mental retardation 1 gene in human tissues. Mol Med Rep 2015; 12:1957-62. [DOI: 10.3892/mmr.2015.3574] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 02/17/2015] [Indexed: 11/06/2022] Open
|
30
|
Pretto DI, Eid JS, Yrigollen CM, Tang HT, Loomis EW, Raske C, Durbin-Johnson B, Hagerman PJ, Tassone F. Differential increases of specific FMR1 mRNA isoforms in premutation carriers. J Med Genet 2014; 52:42-52. [PMID: 25358671 DOI: 10.1136/jmedgenet-2014-102593] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Over 40% of male and ∼16% of female carriers of a premutation FMR1 allele (55-200 CGG repeats) will develop fragile X-associated tremor/ataxia syndrome, an adult onset neurodegenerative disorder, while about 20% of female carriers will develop fragile X-associated primary ovarian insufficiency. Marked elevation in FMR1 mRNA transcript levels has been observed with premutation alleles, and RNA toxicity due to increased mRNA levels is the leading molecular mechanism proposed for these disorders. However, although the FMR1 gene undergoes alternative splicing, it is unknown whether all or only some of the isoforms are overexpressed in premutation carriers and which isoforms may contribute to the premutation pathology. METHODS To address this question, we have applied a long-read sequencing approach using single-molecule real-time (SMRT) sequencing and qRT-PCR. RESULTS Our SMRT sequencing analysis performed on peripheral blood mononuclear cells, fibroblasts and brain tissue samples derived from premutation carriers and controls revealed the existence of 16 isoforms of 24 predicted variants. Although the relative abundance of all mRNA isoforms was significantly increased in the premutation group, as expected based on the bulk increase in mRNA levels, there was a disproportionate (fourfold to sixfold) increase, relative to the overall increase in mRNA, in the abundance of isoforms spliced at both exons 12 and 14, specifically Iso10 and Iso10b, containing the complete exon 15 and differing only in splicing in exon 17. CONCLUSIONS These findings suggest that RNA toxicity may arise from a relative increase of all FMR1 mRNA isoforms. Interestingly, the Iso10 and Iso10b mRNA isoforms, lacking the C-terminal functional sites for fragile X mental retardation protein function, are the most increased in premutation carriers relative to normal, suggesting a functional relevance in the pathology of FMR1-associated disorders.
Collapse
Affiliation(s)
- Dalyir I Pretto
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA
| | - John S Eid
- Pacific Biosciences, Menlo Park, California, USA
| | - Carolyn M Yrigollen
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA
| | - Hiu-Tung Tang
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA
| | - Erick W Loomis
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA
| | - Chris Raske
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA
| | - Blythe Durbin-Johnson
- Department of Public Health Sciences, University of California Davis, School of Medicine, Davis, California, USA
| | - Paul J Hagerman
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA MIND Institute, University of California Davis Medical Center, Sacramento, California, USA
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California, School of Medicine, Davis, California, USA MIND Institute, University of California Davis Medical Center, Sacramento, California, USA
| |
Collapse
|
31
|
Berman RF, Buijsen RA, Usdin K, Pintado E, Kooy F, Pretto D, Pessah IN, Nelson DL, Zalewski Z, Charlet-Bergeurand N, Willemsen R, Hukema RK. Mouse models of the fragile X premutation and fragile X-associated tremor/ataxia syndrome. J Neurodev Disord 2014; 6:25. [PMID: 25136376 PMCID: PMC4135345 DOI: 10.1186/1866-1955-6-25] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 01/29/2014] [Indexed: 11/10/2022] Open
Abstract
Carriers of the fragile X premutation (FPM) have CGG trinucleotide repeat expansions of between 55 and 200 in the 5'-UTR of FMR1, compared to a CGG repeat length of between 5 and 54 for the general population. Carriers were once thought to be without symptoms, but it is now recognized that they can develop a variety of early neurological symptoms as well as being at risk for developing the late onset neurodegenerative disorder fragile X-associated tremor/ataxia syndrome (FXTAS). Several mouse models have contributed to our understanding of FPM and FXTAS, and findings from studies using these models are summarized here. This review also discusses how this information is improving our understanding of the molecular and cellular abnormalities that contribute to neurobehavioral features seen in some FPM carriers and in patients with FXTAS. Mouse models show much of the pathology seen in FPM carriers and in individuals with FXTAS, including the presence of elevated levels of Fmr1 mRNA, decreased levels of fragile X mental retardation protein, and ubiquitin-positive intranuclear inclusions. Abnormalities in dendritic spine morphology in several brain regions are associated with neurocognitive deficits in spatial and temporal memory processes, impaired motor performance, and altered anxiety. In vitro studies have identified altered dendritic and synaptic architecture associated with abnormal Ca(2+) dynamics and electrical network activity. FPM mice have been particularly useful in understanding the roles of Fmr1 mRNA, fragile X mental retardation protein, and translation of a potentially toxic polyglycine peptide in pathology. Finally, the potential for using these and emerging mouse models for preclinical development of therapies to improve neurological function in FXTAS is considered.
Collapse
Affiliation(s)
- Robert F Berman
- Department of Neurological Surgery, Room 502C, UC Davis, 1515 Newton Court, Davis, CA 95618, USA
| | | | - Karen Usdin
- NIDDK, National Institutes of Health, Bethesda, MD, USA
| | | | - Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | | | - Isaac N Pessah
- Department Molecular Biosciences, UC Davis, Davis, CA, USA
| | - David L Nelson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Zachary Zalewski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Rob Willemsen
- Department Clinical Genetics, Erasmus MC, Rotterdam, Netherlands
| | - Renate K Hukema
- Department Clinical Genetics, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
32
|
Taha MS, Nouri K, Milroy LG, Moll JM, Herrmann C, Brunsveld L, Piekorz RP, Ahmadian MR. Subcellular fractionation and localization studies reveal a direct interaction of the fragile X mental retardation protein (FMRP) with nucleolin. PLoS One 2014; 9:e91465. [PMID: 24658146 PMCID: PMC3962360 DOI: 10.1371/journal.pone.0091465] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 02/11/2014] [Indexed: 12/31/2022] Open
Abstract
Fragile X mental Retardation Protein (FMRP) is a well-known regulator of local translation of its mRNA targets in neurons. However, despite its ubiquitous expression, the role of FMRP remains ill-defined in other cell types. In this study we investigated the subcellular distribution of FMRP and its protein complexes in HeLa cells using confocal imaging as well as detergent-free fractionation and size exclusion protocols. We found FMRP localized exclusively to solid compartments, including cytosolic heavy and light membranes, mitochondria, nuclear membrane and nucleoli. Interestingly, FMRP was associated with nucleolin in both a high molecular weight ribosomal and translation-associated complex (≥6 MDa) in the cytosol, and a low molecular weight complex (∼200 kDa) in the nucleoli. Consistently, we identified two functional nucleolar localization signals (NoLSs) in FMRP that are responsible for a strong nucleolar colocalization of the C-terminus of FMRP with nucleolin, and a direct interaction of the N-terminus of FMRP with the arginine-glycine-glycine (RGG) domain of nucleolin. Taken together, we propose a novel mechanism by which a transient nucleolar localization of FMRP underlies a strong nucleocytoplasmic translocation, most likely in a complex with nucleolin and possibly ribosomes, in order to regulate translation of its target mRNAs.
Collapse
Affiliation(s)
- Mohamed S. Taha
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany
| | - Kazem Nouri
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany
| | - Lech G. Milroy
- Laboratory of Chemical Biology and Institute of Complex Molecular Systems, Department of Biomedical Engineering, Technische Universiteit Eindhoven, Eindhoven, the Netherlands
| | - Jens M. Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany
| | - Christian Herrmann
- Department of Physical Chemistry I, Ruhr University Bochum, Bochum, Germany
| | - Luc Brunsveld
- Laboratory of Chemical Biology and Institute of Complex Molecular Systems, Department of Biomedical Engineering, Technische Universiteit Eindhoven, Eindhoven, the Netherlands
| | - Roland P. Piekorz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany
| | - Mohammad R. Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
33
|
Blice-Baum AC, Mihailescu MR. Biophysical characterization of G-quadruplex forming FMR1 mRNA and of its interactions with different fragile X mental retardation protein isoforms. RNA (NEW YORK, N.Y.) 2014; 20:103-114. [PMID: 24249225 PMCID: PMC3866639 DOI: 10.1261/rna.041442.113] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/14/2013] [Indexed: 06/02/2023]
Abstract
Fragile X syndrome, the most common form of inherited mental impairment in humans, is caused by the absence of the fragile X mental retardation protein (FMRP) due to a CGG trinucleotide repeat expansion in the 5'-untranslated region (UTR) and subsequent translational silencing of the fragile x mental retardation-1 (FMR1) gene. FMRP, which is proposed to be involved in the translational regulation of specific neuronal messenger RNA (mRNA) targets, contains an arginine-glycine-glycine (RGG) box RNA binding domain that has been shown to bind with high affinity to G-quadruplex forming mRNA structures. FMRP undergoes alternative splicing, and the binding of FMRP to a proposed G-quadruplex structure in the coding region of its mRNA (named FBS) has been proposed to affect the mRNA splicing events at exon 15. In this study, we used biophysical methods to directly demonstrate the folding of FMR1 FBS into a secondary structure that contains two specific G-quadruplexes and analyze its interactions with several FMRP isoforms. Our results show that minor splice isoforms, ISO2 and ISO3, created by the usage of the second and third acceptor sites at exon 15, bind with higher affinity to FBS than FMRP ISO1, which is created by the usage of the first acceptor site. FMRP ISO2 and ISO3 cannot undergo phosphorylation, an FMRP post-translational modification shown to modulate the protein translation regulation. Thus, their expression has to be tightly regulated, and this might be accomplished by a feedback mechanism involving the FMRP interactions with the G-quadruplex structures formed within FMR1 mRNA.
Collapse
|
34
|
Dury AY, El Fatimy R, Tremblay S, Rose TM, Côté J, De Koninck P, Khandjian EW. Nuclear Fragile X Mental Retardation Protein is localized to Cajal bodies. PLoS Genet 2013; 9:e1003890. [PMID: 24204304 PMCID: PMC3814324 DOI: 10.1371/journal.pgen.1003890] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 09/03/2013] [Indexed: 01/04/2023] Open
Abstract
Fragile X syndrome is caused by loss of function of a single gene encoding the Fragile X Mental Retardation Protein (FMRP). This RNA-binding protein, widely expressed in mammalian tissues, is particularly abundant in neurons and is a component of messenger ribonucleoprotein (mRNP) complexes present within the translational apparatus. The absence of FMRP in neurons is believed to cause translation dysregulation and defects in mRNA transport essential for local protein synthesis and for synaptic development and maturation. A prevalent model posits that FMRP is a nucleocytoplasmic shuttling protein that transports its mRNA targets from the nucleus to the translation machinery. However, it is not known which of the multiple FMRP isoforms, resulting from the numerous alternatively spliced FMR1 transcripts variants, would be involved in such a process. Using a new generation of anti-FMRP antibodies and recombinant expression, we show here that the most commonly expressed human FMRP isoforms (ISO1 and 7) do not localize to the nucleus. Instead, specific FMRP isoforms 6 and 12 (ISO6 and 12), containing a novel C-terminal domain, were the only isoforms that localized to the nuclei in cultured human cells. These isoforms localized to specific p80-coilin and SMN positive structures that were identified as Cajal bodies. The Cajal body localization signal was confined to a 17 amino acid stretch in the C-terminus of human ISO6 and is lacking in a mouse Iso6 variant. As FMRP is an RNA-binding protein, its presence in Cajal bodies suggests additional functions in nuclear post-transcriptional RNA metabolism. Supporting this hypothesis, a missense mutation (I304N), known to alter the KH2-mediated RNA binding properties of FMRP, abolishes the localization of human FMRP ISO6 to Cajal bodies. These findings open unexplored avenues in search for new insights into the pathophysiology of Fragile X Syndrome. Fragile X syndrome is the most common form of inherited mental retardation affecting approximately 1/7000 females and 1/4000 males worldwide. The syndrome is due to the silencing of a single gene, the Fragile Mental Retardation 1 (FMR1), that codes for a protein called the Fragile X mental retardation protein (FMRP). This protein, highly expressed in the brain, controls local protein synthesis essential for neuronal development and maturation. While considerable efforts have been focused on understanding FMRP functions in mental retardation, the pathophysiology of the syndrome is not well understood. Here, we show that in addition to the well-studied roles of FMRP in regulating protein synthesis, a minor species of FMRP different from the major one, is specifically found in structures called Cajal bodies present in the cell nucleus. Our observations suggest that different FMRP species, also called isoforms, might have independent cellular functions. These findings might open new avenues in search for new insights in the pathophysiology of Fragile X Syndrome.
Collapse
Affiliation(s)
- Alain Y. Dury
- Centre de recherche, Institut en santé mentale de Québec, Québec, Québec, Canada
- Département de psychiatrie et des neurosciences, Faculté de médecine, Université Laval, Québec, Québec, Canada
| | - Rachid El Fatimy
- Centre de recherche, Institut en santé mentale de Québec, Québec, Québec, Canada
- Département de psychiatrie et des neurosciences, Faculté de médecine, Université Laval, Québec, Québec, Canada
| | - Sandra Tremblay
- Centre de recherche, Institut en santé mentale de Québec, Québec, Québec, Canada
| | - Timothy M. Rose
- Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine and Center for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| | - Paul De Koninck
- Centre de recherche, Institut en santé mentale de Québec, Québec, Québec, Canada
- Département de Biochimie, Microbiologie et Bio-Informatique, Université Laval, Québec, Québec, Canada
| | - Edouard W. Khandjian
- Centre de recherche, Institut en santé mentale de Québec, Québec, Québec, Canada
- Département de psychiatrie et des neurosciences, Faculté de médecine, Université Laval, Québec, Québec, Canada
- * E-mail:
| |
Collapse
|
35
|
Bagni C, Oostra BA. Fragile X syndrome: From protein function to therapy. Am J Med Genet A 2013; 161A:2809-21. [PMID: 24115651 DOI: 10.1002/ajmg.a.36241] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/28/2013] [Indexed: 12/23/2022]
Abstract
Fragile X syndrome (FXS) is the leading monogenic cause of intellectual disability and autism. The FMR1 gene contains a CGG repeat present in the 5'-untranslated region which can be unstable upon transmission to the next generation. The repeat is up to 55 CGGs long in the normal population. In patients with fragile X syndrome (FXS), a repeat length exceeding 200 CGGs generally leads to methylation of the repeat and the promoter region, which is accompanied by silencing of the FMR1 gene. The disease is a result of lack of expression of the fragile X mental retardation protein leading to severe symptoms, including intellectual disability, hyperactivity, and autistic-like behavior. The FMR1 protein (FMRP) has a number of functions. The translational dysregulation of a subset of mRNAs targeted by FMRP is probably the major contribution to FXS. FMRP is also involved in mRNA transport to synapses where protein synthesis occurs. For some FMRP-bound mRNAs, FMRP is a direct modulator of mRNA stability either by sustaining or preventing mRNA decay. Increased knowledge about the role of FMRP has led to the identification of potential treatments for fragile X syndrome that were often tested first in the different animal models. This review gives an overview about the present knowledge of the function of FMRP and the therapeutic strategies in mouse and man.
Collapse
Affiliation(s)
- Claudia Bagni
- VIB Center for the Biology of Disease, Catholic University of Leuven, Leuven, Belgium; Department of Biomedicine and Prevention, University of Rome, Tor Vergata, Italy
| | | |
Collapse
|
36
|
Comprehensive analysis of the transcriptional landscape of the human FMR1 gene reveals two new long noncoding RNAs differentially expressed in Fragile X syndrome and Fragile X-associated tremor/ataxia syndrome. Hum Genet 2013; 133:59-67. [PMID: 24005575 PMCID: PMC3898532 DOI: 10.1007/s00439-013-1356-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/25/2013] [Indexed: 01/23/2023]
Abstract
The majority of the human genome is transcribed but not translated, giving rise to noncoding RNAs (ncRNAs), including long ncRNAs (lncRNAs, >200 nt) that perform a wide range of functions in gene regulation. The Fragile X mental retardation 1 (FMR1) gene is a microsatellite locus that in the general population contains <55 CGG repeats in its 5′-untranslated region. Expansion of this repeat region to a size of 55-200 CGG repeats, known as premutation, is associated with Fragile X tremor and ataxia syndrome (FXTAS). Further expansion beyond 200 CGG repeats, or full mutation, leads to FMR1 gene silencing and results in Fragile X syndrome (FXS). Using a novel technology called “Deep-RACE”, which combines rapid amplification of cDNA ends (RACE) with next generation sequencing, we systematically interrogated the FMR1 gene locus for the occurrence of novel lncRNAs. We discovered two transcripts, FMR5 and FMR6. FMR5 is a sense lncRNA transcribed upstream of the FMR1 promoter, whereas FMR6 is an antisense transcript overlapping the 3′ region of FMR1. FMR5 was expressed in several human brain regions from unaffected individuals and from full and premutation patients. FMR6 was silenced in full mutation and, unexpectedly, in premutation carriers suggesting abnormal transcription and/or chromatin remodeling prior to transition to the full mutation. These lncRNAs may thus be useful as biomarkers, allowing for early detection and therapeutic intervention in FXS and FXTAS. Finally we show that FMR5 and FMR6 are expressed in peripheral blood leukocytes and propose future studies that correlate lncRNA expression with clinical outcomes.
Collapse
|
37
|
Ferder I, Parborell F, Sundblad V, Chiauzzi V, Gómez K, Charreau EH, Tesone M, Dain L. Expression of fragile X mental retardation protein and Fmr1 mRNA during folliculogenesis in the rat. Reproduction 2013; 145:335-43. [PMID: 23401597 DOI: 10.1530/rep-12-0305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fragile X mental retardation protein (FMRP) belongs to a small family of RNA-binding proteins. Its absence or inactivity is responsible for fragile X syndrome, the most common cause of inherited mental retardation. Despite its ubiquitous expression, FMRP function and expression remain almost understudied in non-neuronal tissues, though previous studies on germline development during oogenesis may suggest a special function of this protein also in ovarian tissue. In addition, the well-documented association of FMR1 premutation state with fragile X-related premature ovarian insufficiency adds interest to the role of FMRP in ovarian physiology. The aim of the present work was to investigate the expression of Fmr1 mRNA and its protein, FMRP, at different stages of rat follicular development. By immunohistochemical studies we demonstrated FMRP expression in granulosa, theca and germ cells in all stages of follicular development. In addition, changes in Fmr1 expression, both at the protein and mRNA levels, were observed. FMRP levels increased upon follicular development while preantral and early antral follicles presented similar levels of Fmr1 transcripts with decreased expression in preovulatory follicles. These observations suggest that Fmr1 expression in the ovary is regulated at different and perhaps independent levels. In addition, our results show expression of at least four different isoforms of FMRP during all stages of follicular growth with expression patterns that differ from those observed in brain and testis. Our study shows a regulated expression of Fmr1, both at mRNA and protein levels, during rat follicular development.
Collapse
Affiliation(s)
- Ianina Ferder
- Instituto de Biología y Medicina Experimental (IByME)- CONICET, Vuelta de Obligado 2490, C1428ADN Ciudad de Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Brackett DM, Qing F, Amieux PS, Sellers DL, Horner PJ, Morris DR. FMR1 transcript isoforms: association with polyribosomes; regional and developmental expression in mouse brain. PLoS One 2013; 8:e58296. [PMID: 23505481 PMCID: PMC3591412 DOI: 10.1371/journal.pone.0058296] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/01/2013] [Indexed: 12/31/2022] Open
Abstract
The primary transcript of the mammalian Fragile X Mental Retardation-1 gene (Fmr1), like many transcripts in the central nervous system, is alternatively spliced to yield mRNAs encoding multiple proteins, which can possess quite different biochemical properties. Despite the fact that the relative levels of the 12 Fmr1 transcript isoforms examined here vary by as much as two orders of magnitude amongst themselves in both adult and embryonic mouse brain, all are associated with polyribosomes, consistent with translation into the corresponding isoforms of the protein product, FMRP (Fragile X Mental Retardation Protein). Employing the RiboTag methodology developed in our laboratory, the relative proportions of the 7 most abundant transcript isoforms were measured specifically in neurons and found to be similar to those identified in whole brain. Measurements of isoform profiles across 11 regions of adult brain yielded similar distributions, with the exceptions of the hippocampus and the olfactory bulb. These two regions differ from most of the brain in relative amounts of transcripts encoding an alternate form of one of the KH RNA binding domains. A possible relationship between patterns of expression in the hippocampus and olfactory bulb and the presence of neuroblasts in these two regions is suggested by the isoform patterns in early embryonic brain and in cultured neural progenitor cells. These results demonstrate that the relative levels of the Fmr1 isoforms are modulated according to developmental stage, highlighting the complex ramifications of losing all the protein isoforms in individuals with Fragile X Syndrome. It should also be noted that, of the eight most prominent FMRP isoforms (1–3, 6–9 and 12) in mouse, only two have the major site of phosphorylation at Ser-499, which is thought to be involved in some of the regulatory interactions of this protein.
Collapse
Affiliation(s)
- David M. Brackett
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Feng Qing
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Paul S. Amieux
- Department of Pharmacology; University of Washington, Seattle, Washington, United States of America
| | - Drew L. Sellers
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States of America
| | - Philip J. Horner
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States of America
| | - David R. Morris
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
39
|
Gareau C, Houssin E, Martel D, Coudert L, Mellaoui S, Huot ME, Laprise P, Mazroui R. Characterization of fragile X mental retardation protein recruitment and dynamics in Drosophila stress granules. PLoS One 2013; 8:e55342. [PMID: 23408971 PMCID: PMC3567066 DOI: 10.1371/journal.pone.0055342] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 12/21/2012] [Indexed: 01/27/2023] Open
Abstract
The RNA-binding protein Fragile X Mental Retardation (FMRP) is an evolutionarily conserved protein that is particularly abundant in the brain due to its high expression in neurons. FMRP deficiency causes fragile X mental retardation syndrome. In neurons, FMRP controls the translation of target mRNAs in part by promoting dynamic transport in and out neuronal RNA granules. We and others have previously shown that upon stress, mammalian FMRP dissociates from translating polysomes to localize into neuronal-like granules termed stress granules (SG). This localization of FMRP in SG is conserved in Drosophila. Whether FMRP plays a key role in SG formation, how FMRP is recruited into SG, and whether its association with SG is dynamic are currently unknown. In contrast with mammalian FMRP, which has two paralog proteins, Drosophila FMR1 (dFMRP) is encoded by a single gene that has no paralog. Using this genetically simple model, we assessed the role of dFMRP in SG formation and defined the determinants required for its recruitment in SG as well as its dynamics in SG. We show that dFMRP is dispensable for SG formation in vitro and ex vivo. FRAP experiments showed that dFMRP shuttles in and out SG. The shuttling activity of dFMRP is mediated by a protein-protein interaction domain located at the N-terminus of the protein. This domain is, however, dispensable for the localization of dFMRP in SG. This localization of dFMRP in SG requires the KH and RGG motifs which are known to mediate RNA binding, as well as the C-terminal glutamine/asparagine rich domain. Our studies thus suggest that the mechanisms controlling the recruitment of FMRP into SG and those that promote its shuttling between granules and the cytosol are uncoupled. To our knowledge, this is the first demonstration of the regulated shuttling activity of a SG component between RNA granules and the cytosol.
Collapse
Affiliation(s)
- Cristina Gareau
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - Elise Houssin
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - David Martel
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - Laetitia Coudert
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - Samia Mellaoui
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - Marc-Etienne Huot
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - Patrick Laprise
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
| | - Rachid Mazroui
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, Centre de recherche le CHU de Quebec, Quebec, Canada
- * E-mail:
| |
Collapse
|
40
|
Bagni C, Tassone F, Neri G, Hagerman R. Fragile X syndrome: causes, diagnosis, mechanisms, and therapeutics. J Clin Invest 2012. [PMID: 23202739 DOI: 10.1172/jci63141] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fragile X syndrome (FXS) is the most frequent form of inherited intellectual disability and is also linked to other neurologic and psychiatric disorders. FXS is caused by a triplet expansion that inhibits expression of the FMR1 gene; the gene product, FMRP, regulates mRNA metabolism in the brain and thus controls the expression of key molecules involved in receptor signaling and spine morphology. While there is no definitive cure for FXS, the understanding of FMRP function has paved the way for rational treatment designs that could potentially reverse many of the neurobiological changes observed in FXS. Additionally, behavioral, pharmacological, and cognitive interventions can raise the quality of life for both patients and their families.
Collapse
Affiliation(s)
- Claudia Bagni
- Katholieke Universiteit Leuven, Center for Human Genetics, Leuven, Belgium.
| | | | | | | |
Collapse
|
41
|
Gareau C, Martel D, Coudert L, Mellaoui S, Mazroui R. Characterization of Fragile X Mental Retardation Protein granules formation and dynamics in Drosophila. Biol Open 2012; 2:68-81. [PMID: 23336078 PMCID: PMC3545270 DOI: 10.1242/bio.20123012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 10/03/2012] [Indexed: 01/30/2023] Open
Abstract
FMRP is an evolutionarily conserved protein that is highly expressed in neurons and its deficiency causes fragile X mental retardation syndrome. FMRP controls the translation of target mRNAs in part by promoting their dynamic transport in neuronal RNA granules. We have previously shown that high expression of mammalian FMRP induces formation of granules termed FMRP granules. These RNA granules are reminiscent of neuronal granules, of stress granules, as well as of the recently described in vitro-assembled granules. In contrast with mammalian FMRP, which has two paralog proteins, Drosophila FMRP (dFMRP) is encoded by a single gene that has no paralog. Using this genetically simple organism, we investigated formation and dynamics of FMRP granules. We found that increased expression of dFMRP in Drosophila cells induces the formation of dynamic dFMRP RNA granules. Mutagenesis studies identified the N-terminal protein–protein domain of dFMRP as a key determinant for FMRP granules formation. The RGG RNA binding motif of dFMRP is dispensable for dFMRP granules formation since its deletion does not prevent formation of those granules. Deletion of the RGG motif reduced, however, dFMRP trafficking between FMRP granules and the cytosol. Similarly, deletion of a large part of the KH RNA binding motif of dFMRP had no effect on formation of dFMRP-granules, but diminished the shuttling activity of dFMRP. Our results thus suggest that the mechanisms controlling formation of RNA granules and those promoting their dynamics are uncoupled. This study opens new avenues to further elucidate the molecular mechanisms controlling FMRP trafficking with its associated mRNAs in and out of RNA granules.
Collapse
Affiliation(s)
- Cristina Gareau
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Laval University, CHUQ Research Center/St-François d'Assise Research Center , Quebec, QC G1L 3L5 , Canada
| | | | | | | | | |
Collapse
|
42
|
Jeon SJ, Han SH, Yang SI, Choi JW, Kwon KJ, Park SH, Kim HY, Cheong JH, Ryu JH, Ko KH, Wells DG, Shin CY. Positive feedback regulation of Akt-FMRP pathway protects neurons from cell death. J Neurochem 2012; 123:226-38. [DOI: 10.1111/j.1471-4159.2012.07886.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
43
|
Evans TL, Blice-Baum AC, Mihailescu MR. Analysis of the Fragile X mental retardation protein isoforms 1, 2 and 3 interactions with the G-quadruplex forming semaphorin 3F mRNA. ACTA ACUST UNITED AC 2012; 8:642-9. [DOI: 10.1039/c1mb05322a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
44
|
Molecular and Cellular Aspects of Mental Retardation in the Fragile X Syndrome: From Gene Mutation/s to Spine Dysmorphogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:517-51. [DOI: 10.1007/978-3-7091-0932-8_23] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
45
|
Bardoni B, Abekhoukh S, Zongaro S, Melko M. Intellectual disabilities, neuronal posttranscriptional RNA metabolism, and RNA-binding proteins: three actors for a complex scenario. PROGRESS IN BRAIN RESEARCH 2012; 197:29-51. [PMID: 22541287 DOI: 10.1016/b978-0-444-54299-1.00003-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intellectual disability (ID) is the most frequent cause of serious handicap in children and young adults and interests 2-3% of worldwide population, representing a serious problem from the medical, social, and economic points of view. The causes are very heterogeneous. Genes involved in ID have various functions altering different pathways important in neuronal function. Regulation of mRNA metabolism is particularly important in neurons for synaptic structure and function. Here, we review ID due to alteration of mRNA metabolism. Functional absence of some RNA-binding proteins--namely, FMRP, FMR2P, PQBP1, UFP3B, VCX-A--causes different forms of ID. These proteins are involved in different steps of RNA metabolism and, even if a detailed analysis of their RNA targets has been performed so far only for FMRP, it appears clear that they modulate some aspects (translation, stability, transport, and sublocalization) of a subset of RNAs coding for proteins, whose function must be relevant for neurons. Two other proteins, DYRK1A and CDKL5, involved in Down syndrome and Rett syndrome, respectively, have been shown to have an impact on splicing efficiency of specific mRNAs. Both proteins are kinases and their effect is indirect. Interestingly, both are localized in nuclear speckles, the nuclear domains where splicing factors are assembled, stocked, and recycled and influence their biogenesis and/or their organization.
Collapse
Affiliation(s)
- Barbara Bardoni
- Institute of Molecular and Cellular Pharmacology, CNRS-UMR6097, Université de Nice Sophia-Antipolis,Valbonne, France.
| | | | | | | |
Collapse
|
46
|
Winograd C, Ceman S. Fragile X family members have important and non-overlapping functions. Biomol Concepts 2011; 2:343-52. [DOI: 10.1515/bmc.2011.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 06/29/2011] [Indexed: 01/15/2023] Open
Abstract
AbstractThe fragile X family of genes encodes a small family of RNA binding proteins including FMRP, FXR1P and FXR2P that were identified in the 1990s. All three members are encoded by 17 exons and show alternative splicing at the 3′ ends of their respective transcripts. They share significant homology in the protein functional domains, including the Tudor domains, the nuclear localization sequence, a protein-protein interaction domain, the KH1 and KH2 domains and the nuclear export sequence. Fragile X family members are found throughout the animal kingdom, although all three members are not consistently present in species outside of mammals: only two family members are present in the avian species examined, Gallus gallus and Taeniopygia guttata, and in the frog Xenopus tropicalis. Although present in many tissues, the functions of the fragile X family members differ, which are particularly evident in knockout studies performed in animals. The fragile X family members play roles in normal neuronal function and in the case of FXR1, in muscle function.
Collapse
Affiliation(s)
- Claudia Winograd
- 2Neuroscience Program and College of Medicine, University of Illinois, 601 S. Goodwin Avenue, Urbana–Champaign, IL 61801, USA
| | | |
Collapse
|
47
|
Pirozzi F, Tabolacci E, Neri G. The FRAXopathies: definition, overview, and update. Am J Med Genet A 2011; 155A:1803-16. [PMID: 21739597 DOI: 10.1002/ajmg.a.34113] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 04/22/2011] [Indexed: 01/12/2023]
Abstract
The fragile X syndrome, fragile X tremor ataxia syndrome, and premature ovarian insufficiency are conditions related to the X chromosome folate-sensitive fragile site FRAXA. Therefore, we propose that they are considered as a family of disorders under the general designation of FRAXopathies. The present review will outline the main clinical and molecular features of these disorders, with special emphasis on the pathogenic mechanisms that lead to distinct phenotypes, starting from related mutations. The understanding of these mechanisms is already generating promising therapeutic approaches.
Collapse
|
48
|
Fatemi SH, Folsom TD. Dysregulation of fragile × mental retardation protein and metabotropic glutamate receptor 5 in superior frontal cortex of individuals with autism: a postmortem brain study. Mol Autism 2011; 2:6. [PMID: 21548960 PMCID: PMC3488976 DOI: 10.1186/2040-2392-2-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 05/06/2011] [Indexed: 12/26/2022] Open
Abstract
Background Fragile X syndrome is caused by loss of function of the fragile X mental retardation 1 (FMR1) gene and shares multiple phenotypes with autism. We have previously found reduced expression of the protein product of FMR1 (FMRP) in vermis of adults with autism. Methods In the current study, we have investigated levels of FMRP in the superior frontal cortex of people with autism and matched controls using Western blot analysis. Because FMRP regulates the translation of multiple genes, we also measured protein levels for downstream molecules metabotropic glutamate receptor 5 (mGluR5) and γ-aminobutyric acid (GABA) A receptor β3 (GABRβ3), as well as glial fibrillary acidic protein (GFAP). Results We observed significantly reduced levels of protein for FMRP in adults with autism, significantly increased levels of protein for mGluR5 in children with autism and significantly increased levels of GFAP in adults and children with autism. We found no change in expression of GABRβ3. Our results for FMRP, mGluR5 and GFAP confirm our previous work in the cerebellar vermis of people with autism. Conclusion These changes may be responsible for cognitive deficits and seizure disorder in people with autism.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Division of Neuroscience Research, Department of Psychiatry, University of Minnesota Medical School, 420 Delaware Street SE, MMC 392, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
49
|
Tassone F, De Rubeis S, Carosi C, La Fata G, Serpa G, Raske C, Willemsen R, Hagerman PJ, Bagni C. Differential usage of transcriptional start sites and polyadenylation sites in FMR1 premutation alleles. Nucleic Acids Res 2011; 39:6172-85. [PMID: 21478165 PMCID: PMC3152321 DOI: 10.1093/nar/gkr100] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
5′- and 3′-untranslated regions (UTRs) are important regulators of gene expression and play key roles in disease progression and susceptibility. The 5′-UTR of the fragile X mental retardation 1 (FMR1) gene contains a CGG repeat element that is expanded (>200 CGG repeats; full mutation) and methylated in fragile X syndrome (FXS), the most common form of inherited intellectual disability (ID) and known cause of autism. Significant phenotypic involvement has also emerged in some individuals with the premutation (55–200 CGG repeats), including fragile X-associated premature ovarian insufficiency (FXPOI) in females, and the neurodegenerative disorder, fragile X-associated tremor/ataxia syndrome (FXTAS), in older adult carriers. Here, we show that FMR1 mRNA in human and mouse brain is expressed as a combination of multiple isoforms that use alternative transcriptional start sites and different polyadenylation sites. Furthermore, we have identified a novel human transcription start site used in brain but not in lymphoblastoid cells, and have detected FMR1 isoforms generated through the use of both canonical and non-canonical polyadenylation signals. Importantly, in both human and mouse, a specific regulation of the UTRs is observed in brain of FMR1 premutation alleles, suggesting that the transcript variants may play a role in premutation-related pathologies.
Collapse
Affiliation(s)
- Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
- *To whom correspondence should be addressed. Tel: +39 06 72596063/+32 16330944; Fax: +39 06 72596058/+39 16330939; ;
| | - Silvia De Rubeis
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Chiara Carosi
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Giorgio La Fata
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Gisele Serpa
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Christopher Raske
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Rob Willemsen
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Paul J. Hagerman
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
| | - Claudia Bagni
- Department of Biochemistry and Molecular Medicine, University of California, Davis School of Medicine, Davis, CA, USA M.I.N.D. Institute, University of California, Davis Medical Center, Sacramento, CA, USA, Center for Human Genetics, Katholieke Universiteit Leuven, Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium, Fondazione Santa Lucia, IRCCS, Rome, Italy, Genomic Engineering Group, Chemical and Food Engineering Department, Federal University of Santa Catarina, Florianópolis, Brazil, CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands and Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Rome, Italy
- *To whom correspondence should be addressed. Tel: +39 06 72596063/+32 16330944; Fax: +39 06 72596058/+39 16330939; ;
| |
Collapse
|
50
|
Smith RM, Sadee W. Synaptic signaling and aberrant RNA splicing in autism spectrum disorders. Front Synaptic Neurosci 2011; 3:1. [PMID: 21423409 PMCID: PMC3059609 DOI: 10.3389/fnsyn.2011.00001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Accepted: 01/12/2011] [Indexed: 11/13/2022] Open
Abstract
Interactions between presynaptic and postsynaptic cellular adhesion molecules (CAMs) drive synapse maturation during development. These trans-synaptic interactions are regulated by alternative splicing of CAM RNAs, which ultimately determines neurotransmitter phenotype. The diverse assortment of RNAs produced by alternative splicing generates countless protein isoforms necessary for guiding specialized cell-to-cell connectivity. Failure to generate the appropriate synaptic adhesion proteins is associated with disrupted glutamatergic and gamma-aminobutyric acid signaling, resulting in loss of activity-dependent neuronal plasticity, and risk for developmental disorders, including autism. While the majority of genetic mutations currently linked to autism are rare variants that change the protein-coding sequence of synaptic candidate genes, regulatory polymorphisms affecting constitutive and alternative splicing have emerged as risk factors in numerous other diseases, accounting for an estimated 40–60% of general disease risk. Here, we review the relationship between aberrant RNA splicing of synapse-related genes and autism spectrum disorders.
Collapse
Affiliation(s)
- Ryan M Smith
- Program in Pharmacogenomics, Department of Pharmacology, The Ohio State University Columbus, OH, USA
| | | |
Collapse
|