193 results on '"Usdin, K."'
Search Results
52. Activation Ratio Correlates with IQ in Female Carriers of the FMR1 Premutation.
- Author
-
Protic D, Polli R, Hwang YH, Mendoza G, Hagerman R, Durbin-Johnson B, Hayward BE, Usdin K, Murgia A, and Tassone F
- Subjects
- Female, Animals, Reproducibility of Results, Heterozygote, Methylation, Alleles, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism
- Abstract
Carriers of the FMR1 premutation (PM) allele are at risk of one or more clinical conditions referred to as FX premutation-associated conditions (FXPAC). Since the FMR1 gene is on the X chromosome, the activation ratio (AR) may impact the risk, age of onset, progression, and severity of these conditions. The aim of this study was to evaluate the reliability of AR measured using different approaches and to investigate potential correlations with clinical outcomes. Molecular and clinical assessments were obtained for 30 PM female participants, and AR was assessed using both Southern blot analysis (AR-Sb) and methylation PCR (AR-mPCR). Higher ARs were associated with lower FMR1 transcript levels for any given repeat length. The higher AR-Sb was significantly associated with performance, verbal, and full-scale IQ scores, confirming previous reports. However, the AR-mPCR was not significantly associated ( p > 0.05) with these measures. Similarly, the odds of depression and the number of medical conditions were correlated with higher AR-Sb but not correlated with a higher AR-mPCR. This study suggests that AR-Sb may be a more reliable measure of the AR in female carriers of PM alleles. However, further studies are warranted in a larger sample size to fully evaluate the methylation status in these participants and how it may affect the clinical phenotype.
- Published
- 2023
- Full Text
- View/download PDF
53. Clinical implications of somatic allele expansion in female FMR1 premutation carriers.
- Author
-
Aishworiya R, Hwang YH, Santos E, Hayward B, Usdin K, Durbin-Johnson B, Hagerman R, and Tassone F
- Subjects
- Alleles, Adult, Young Adult, Fragile X Mental Retardation Protein genetics, Child, Preschool, RNA, Messenger, Female, Humans, Aged, Ataxia, Middle Aged, Aged, 80 and over, Adolescent, Infant, Child, Tremor, Trinucleotide Repeat Expansion, Fragile X Syndrome genetics
- Abstract
Carriers of a premutation allele (PM) in the FMR1 gene are at risk of developing a number of Fragile X premutation asssociated disorders (FXPAC), including Fragile X-associated Tremor/Ataxia Syndrome (FXTAS), Fragile X-associated Primary Ovarian Insufficiency (FXPOI), and Fragile X-associated neuropsychiatric disorders (FXAND). We have recently reported somatic CGG allele expansion in female PM; however, its clinical significance remains unclear. The aim of this study was to examine the potential clinical association between somatic FMR1 allele instability and PM associated disorders. Participants comprised of 424 female PM carriers age 0.3- 90 years. FMR1 molecular measures and clinical information on the presence of medical conditions, were determined for all subjects for primary analysis. Two sub-groups of participants (age ≥ 25, N = 377 and age ≥ 50, N = 134) were used in the analysis related to presence of FXPOI and FXTAS, respectively. Among all participants (N = 424), the degree of instability (expansion) was significantly higher (median 2.5 vs 2.0, P = 0.026) in participants with a diagnosis of attention deficit hyperactivity disorder (ADHD) compared to those without. FMR1 mRNA expression was significantly higher in subjects with any psychiatric disorder diagnosis (P = 0.0017); specifically, in those with ADHD (P = 0.009), and with depression (P = 0.025). Somatic FMR1 expansion was associated with the presence of ADHD in female PM and FMR1 mRNA levels were associated with the presence of mental health disorders. The findings of our research are innovative as they suggest a potential role of the CGG expansion in the clinical phenotype of PM and may potentially guide clinical prognosis and management., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
54. Repeat expansions nested within tandem CNVs: a unique structural change in GLS exemplifies the diagnostic challenges of non-coding pathogenic variation.
- Author
-
Fazal S, Danzi MC, van Kuilenburg ABP, Reich S, Traschütz A, Bender B, Leen R, Toro C, Usdin K, Hayward B, Adams DR, van Karnebeek CDM, Ferreira CR, D'Sousa P, Network UD, Tekin M, Züchner S, and Synofzik M
- Subjects
- Humans, 5' Untranslated Regions, Ataxia diagnosis, Ataxia genetics, Glutaminase genetics
- Abstract
Glutaminase deficiency has recently been associated with ataxia and developmental delay due to repeat expansions in the 5'UTR of the glutaminase (GLS) gene. Patients with the described GLS repeat expansion may indeed remain undiagnosed due to the rarity of this variant, the challenge of its detection and the recency of its discovery. In this study, we combined advanced bioinformatics screening of ~3000 genomes and ~1500 exomes with optical genome mapping and long-read sequencing for confirmation studies. We identified two GLS families, previously intensely and unsuccessfully analyzed. One family carries an unusual and complex structural change involving a homozygous repeat expansion nested within a quadruplication event in the 5'UTR of GLS. Glutaminase deficiency and its metabolic consequences were validated by in-depth biochemical analysis. The identified GLS patients showed progressive early-onset ataxia, cognitive deficits, pyramidal tract damage and optic atrophy, thus demonstrating susceptibility of several specific neuron populations to glutaminase deficiency. This large-scale screening study demonstrates the ability of bioinformatics analysis-validated by latest state-of-the-art technologies (optical genome mapping and long-read sequencing)-to effectively flag complex repeat expansions using short-read datasets and thus facilitate diagnosis of ultra-rare disorders., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
55. S1-END-seq reveals DNA secondary structures in human cells.
- Author
-
Matos-Rodrigues G, van Wietmarschen N, Wu W, Tripathi V, Koussa NC, Pavani R, Nathan WJ, Callen E, Belinky F, Mohammed A, Napierala M, Usdin K, Ansari AZ, Mirkin SM, and Nussenzweig A
- Subjects
- DNA metabolism, DNA Breaks, Double-Stranded, DNA Replication, Humans, Nucleic Acid Conformation, DNA, Cruciform, Nylons
- Abstract
DNA becomes single stranded (ssDNA) during replication, transcription, and repair. Transiently formed ssDNA segments can adopt alternative conformations, including cruciforms, triplexes, and quadruplexes. To determine whether there are stable regions of ssDNA in the human genome, we utilized S1-END-seq to convert ssDNA regions to DNA double-strand breaks, which were then processed for high-throughput sequencing. This approach revealed two predominant non-B DNA structures: cruciform DNA formed by expanded (TA)
n repeats that accumulate in microsatellite unstable human cancer cell lines and DNA triplexes (H-DNA) formed by homopurine/homopyrimidine mirror repeats common across a variety of cell lines. We show that H-DNA is enriched during replication, that its genomic location is highly conserved, and that H-DNA formed by (GAA)n repeats can be disrupted by treatment with a (GAA)n -binding polyamide. Finally, we show that triplex-forming repeats are hotspots for mutagenesis. Our results identify dynamic DNA secondary structures in vivo that contribute to elevated genome instability., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
56. Mismatch repair is a double-edged sword in the battle against microsatellite instability.
- Author
-
Miller CJ and Usdin K
- Subjects
- Animals, Genomic Instability, Humans, Mammals genetics, Microsatellite Repeats, DNA Mismatch Repair genetics, Microsatellite Instability
- Abstract
Roughly 3% of the human genome consists of microsatellites or tracts of short tandem repeats (STRs). These STRs are often unstable, undergoing high-frequency expansions (increases) or contractions (decreases) in the number of repeat units. Some microsatellite instability (MSI) is seen at multiple STRs within a single cell and is associated with certain types of cancer. A second form of MSI is characterised by expansion of a single gene-specific STR and such expansions are responsible for a group of 40+ human genetic disorders known as the repeat expansion diseases (REDs). While the mismatch repair (MMR) pathway prevents genome-wide MSI, emerging evidence suggests that some MMR factors are directly involved in generating expansions in the REDs. Thus, MMR suppresses some forms of expansion while some MMR factors promote expansion in other contexts. This review will cover what is known about the paradoxical effect of MMR on microsatellite expansion in mammalian cells.
- Published
- 2022
- Full Text
- View/download PDF
57. Both cis and trans-acting genetic factors drive somatic instability in female carriers of the FMR1 premutation.
- Author
-
Hwang YH, Hayward BE, Zafarullah M, Kumar J, Durbin Johnson B, Holmans P, Usdin K, and Tassone F
- Subjects
- 5' Untranslated Regions, Alleles, Ataxia, Child, Female, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Humans, Mutation, Trans-Activators genetics, Tremor, Trinucleotide Repeat Expansion, Fragile X Syndrome genetics, Intellectual Disability genetics
- Abstract
The fragile X mental retardation (FMR1) gene contains an expansion-prone CGG repeat within its 5' UTR. Alleles with 55-200 repeats are known as premutation (PM) alleles and confer risk for one or more of the FMR1 premutation (PM) disorders that include Fragile X-associated Tremor/Ataxia Syndrome (FXTAS), Fragile X-associated Primary Ovarian Insufficiency (FXPOI), and Fragile X-Associated Neuropsychiatric Disorders (FXAND). PM alleles expand on intergenerational transmission, with the children of PM mothers being at risk of inheriting alleles with > 200 CGG repeats (full mutation FM) alleles) and thus developing Fragile X Syndrome (FXS). PM alleles can be somatically unstable. This can lead to individuals being mosaic for multiple size alleles. Here, we describe a detailed evaluation of somatic mosaicism in a large cohort of female PM carriers and show that 94% display some evidence of somatic instability with the presence of a series of expanded alleles that differ from the next allele by a single repeat unit. Using two different metrics for instability that we have developed, we show that, as with intergenerational instability, there is a direct relationship between the extent of somatic expansion and the number of CGG repeats in the originally inherited allele and an inverse relationship with the number of AGG interruptions. Expansions are progressive as evidenced by a positive correlation with age and by examination of blood samples from the same individual taken at different time points. Our data also suggests the existence of other genetic or environmental factors that affect the extent of somatic expansion. Importantly, the analysis of candidate single nucleotide polymorphisms (SNPs) suggests that two DNA repair factors, FAN1 and MSH3, may be modifiers of somatic expansion risk in the PM population as observed in other repeat expansion disorders., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
58. Stool is a sensitive and noninvasive source of DNA for monitoring expansion in repeat expansion disease mouse models.
- Author
-
Zhao X, McHugh C, Coffey SR, Jimenez DA, Adams E, Carroll JB, and Usdin K
- Subjects
- Animals, Central Nervous System, DNA, Disease Models, Animal, Mice, Trinucleotide Repeat Expansion genetics, Huntington Disease genetics, Spinocerebellar Ataxias
- Abstract
Repeat expansion diseases are a large group of human genetic disorders caused by expansion of a specific short tandem repeat tract. Expansion in somatic cells affects age of onset and disease severity in some of these disorders. However, alleles in DNA derived from blood, a commonly used source of DNA, usually show much less expansion than disease-relevant cells in the central nervous system in both humans and mouse models. Here we examined the extent of expansion in different DNA sources from mouse models of the fragile X-related disorders, Huntington's disease, spinocerebellar ataxia type 1 and spinocerebellar ataxia type 2. We found that DNA isolated from stool is a much better indicator of somatic expansion than DNA from blood. As stool is a sensitive and noninvasive source of DNA, it can be useful for studies of factors affecting the risk of expansion, or the monitoring of treatments aimed at reducing expansion in preclinical trials, as it would allow expansions to be examined longitudinally in the same animal and allow significant changes in expansion to be observed much earlier than is possible with other DNA sources., Competing Interests: Competing interests J.B.C. is on the scientific advisory board, and has shares in Triplet Therapeutics, which is a company focused on modulating somatic instability in HD and other diseases. Other authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
59. FAN1's protection against CGG repeat expansion requires its nuclease activity and is FANCD2-independent.
- Author
-
Zhao X, Lu H, and Usdin K
- Subjects
- Animals, DNA Repair Enzymes metabolism, Endodeoxyribonucleases chemistry, Endodeoxyribonucleases genetics, Exodeoxyribonucleases chemistry, Exodeoxyribonucleases genetics, Fanconi Anemia Complementation Group D2 Protein genetics, Fanconi Anemia Complementation Group D2 Protein metabolism, Mice, Mice, Inbred C57BL, Multifunctional Enzymes chemistry, Multifunctional Enzymes genetics, MutS Homolog 3 Protein metabolism, Point Mutation, Catalytic Domain, Endodeoxyribonucleases metabolism, Exodeoxyribonucleases metabolism, Multifunctional Enzymes metabolism, Trinucleotide Repeat Expansion
- Abstract
The Repeat Expansion Diseases, a large group of human diseases that includes the fragile X-related disorders (FXDs) and Huntington's disease (HD), all result from expansion of a disease-specific microsatellite via a mechanism that is not fully understood. We have previously shown that mismatch repair (MMR) proteins are required for expansion in a mouse model of the FXDs, but that the FANCD2 and FANCI associated nuclease 1 (FAN1), a component of the Fanconi anemia (FA) DNA repair pathway, is protective. FAN1's nuclease activity has been reported to be dispensable for protection against expansion in an HD cell model. However, we show here that in a FXD mouse model a point mutation in the nuclease domain of FAN1 has the same effect on expansion as a null mutation. Furthermore, we show that FAN1 and another nuclease, EXO1, have an additive effect in protecting against MSH3-dependent expansions. Lastly, we show that the loss of FANCD2, a vital component of the Fanconi anemia DNA repair pathway, has no effect on expansions. Thus, FAN1 protects against MSH3-dependent expansions without diverting the expansion intermediates into the canonical FA pathway and this protection depends on FAN1 having an intact nuclease domain., (Published by Oxford University Press on behalf of Nucleic Acids Research 2021.)
- Published
- 2021
- Full Text
- View/download PDF
60. Mechanisms of Genome Instability in the Fragile X-Related Disorders.
- Author
-
Hayward BE and Usdin K
- Subjects
- Aneuploidy, Ataxia physiopathology, DNA Repeat Expansion genetics, DNA Replication genetics, Female, Fragile X Syndrome physiopathology, Genomic Instability genetics, Humans, Primary Ovarian Insufficiency physiopathology, Tremor physiopathology, Trinucleotide Repeat Expansion genetics, Ataxia genetics, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Primary Ovarian Insufficiency genetics, Tremor genetics
- Abstract
The Fragile X-related disorders (FXDs), which include the intellectual disability fragile X syndrome (FXS), are disorders caused by expansion of a CGG-repeat tract in the 5' UTR of the X-linked FMR1 gene. These disorders are named for FRAXA, the folate-sensitive fragile site that localizes with the CGG-repeat in individuals with FXS. Two pathological FMR1 allele size classes are distinguished. Premutation (PM) alleles have 54-200 repeats and confer the risk of fragile X-associated tremor/ataxia syndrome (FXTAS) and fragile X-associated primary ovarian insufficiency (FXPOI). PM alleles are prone to both somatic and germline expansion, with female PM carriers being at risk of having a child with >200+ repeats. Inheritance of such full mutation (FM) alleles causes FXS. Contractions of PM and FM alleles can also occur. As a result, many carriers are mosaic for different sized alleles, with the clinical presentation depending on the proportions of these alleles in affected tissues. Furthermore, it has become apparent that the chromosomal fragility of FXS individuals reflects an underlying problem that can lead to chromosomal numerical and structural abnormalities. Thus, large numbers of CGG-repeats in the FMR1 gene predisposes individuals to multiple forms of genome instability. This review will discuss our current understanding of these processes.
- Published
- 2021
- Full Text
- View/download PDF
61. Common Threads: Aphidicolin-Inducible and Folate-Sensitive Fragile Sites in the Human Genome.
- Author
-
Lokanga RA, Kumari D, and Usdin K
- Abstract
The human genome has many chromosomal regions that are fragile, demonstrating chromatin breaks, gaps, or constrictions on exposure to replication stress. Common fragile sites (CFSs) are found widely distributed in the population, with the largest subset of these sites being induced by aphidicolin (APH). Other fragile sites are only found in a subset of the population. One group of these so-called rare fragile sites (RFSs) is induced by folate stress. APH-inducible CFSs are generally located in large transcriptionally active genes that are A + T rich and often enriched for tracts of AT-dinucleotide repeats. In contrast, all the folate-sensitive sites mapped to date consist of transcriptionally silenced CGG microsatellites. Thus, all the folate-sensitive fragile sites may have a very similar molecular basis that differs in key ways from that of the APH CFSs. The folate-sensitive FSs include FRAXA that is associated with Fragile X syndrome (FXS), the most common heritable form of intellectual disability. Both CFSs and RFSs can cause chromosomal abnormalities. Recent work suggests that both APH-inducible fragile sites and FRAXA undergo Mitotic DNA synthesis (MiDAS) when exposed to APH or folate stress, respectively. Interestingly, blocking MiDAS in both cases prevents chromosome fragility but increases the risk of chromosome mis-segregation. MiDAS of both APH-inducible and FRAXA involves conservative DNA replication and POLD3, an accessory subunit of the replicative polymerase Pol δ that is essential for break-induced replication (BIR). Thus, MiDAS is thought to proceed via some form of BIR-like process. This review will discuss the recent work that highlights the similarities and differences between these two groups of fragile sites and the growing evidence for the presence of many more novel fragile sites in the human genome., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Lokanga, Kumari and Usdin.)
- Published
- 2021
- Full Text
- View/download PDF
62. (Dys)function Follows Form: Nucleic Acid Structure, Repeat Expansion, and Disease Pathology in FMR1 Disorders.
- Author
-
Zhao X and Usdin K
- Subjects
- Animals, Fragile X Mental Retardation Protein chemistry, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome metabolism, Fragile X Syndrome pathology, Humans, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Trinucleotide Repeat Expansion
- Abstract
Fragile X-related disorders (FXDs), also known as FMR1 disorders, are examples of repeat expansion diseases (REDs), clinical conditions that arise from an increase in the number of repeats in a disease-specific microsatellite. In the case of FXDs, the repeat unit is CGG/CCG and the repeat tract is located in the 5' UTR of the X-linked FMR1 gene. Expansion can result in neurodegeneration, ovarian dysfunction, or intellectual disability depending on the number of repeats in the expanded allele. A growing body of evidence suggests that the mutational mechanisms responsible for many REDs share several common features. It is also increasingly apparent that in some of these diseases the pathologic consequences of expansion may arise in similar ways. It has long been known that many of the disease-associated repeats form unusual DNA and RNA structures. This review will focus on what is known about these structures, the proteins with which they interact, and how they may be related to the causative mutation and disease pathology in the FMR1 disorders.
- Published
- 2021
- Full Text
- View/download PDF
63. Editorial: Proceedings of the "Fourth International Conference of the FMR1 Premutation: Basic Mechanisms, Clinical Involvement and Therapy".
- Author
-
Usdin K, Rodriguez-Revenga L, Willemsen R, Hukema R, and Giulivi C
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
64. Modifiers of Somatic Repeat Instability in Mouse Models of Friedreich Ataxia and the Fragile X-Related Disorders: Implications for the Mechanism of Somatic Expansion in Huntington's Disease.
- Author
-
Zhao X, Kumari D, Miller CJ, Kim GY, Hayward B, Vitalo AG, Pinto RM, and Usdin K
- Subjects
- Animals, Humans, Mice, DNA Mismatch Repair genetics, Fragile X Syndrome genetics, Friedreich Ataxia genetics, Genes, Modifier genetics, Genomic Instability genetics, Huntington Disease genetics, Trinucleotide Repeat Expansion genetics
- Abstract
Huntington's disease (HD) is one of a large group of human disorders that are caused by expanded DNA repeats. These repeat expansion disorders can have repeat units of different size and sequence that can be located in any part of the gene and, while the pathological consequences of the expansion can differ widely, there is evidence to suggest that the underlying mutational mechanism may be similar. In the case of HD, the expanded repeat unit is a CAG trinucleotide located in exon 1 of the huntingtin (HTT) gene, resulting in an expanded polyglutamine tract in the huntingtin protein. Expansion results in neuronal cell death, particularly in the striatum. Emerging evidence suggests that somatic CAG expansion, specifically expansion occurring in the brain during the lifetime of an individual, contributes to an earlier disease onset and increased severity. In this review we will discuss mouse models of two non-CAG repeat expansion diseases, specifically the Fragile X-related disorders (FXDs) and Friedreich ataxia (FRDA). We will compare and contrast these models with mouse and patient-derived cell models of various other repeat expansion disorders and the relevance of these findings for somatic expansion in HD. We will also describe additional genetic factors and pathways that modify somatic expansion in the FXD mouse model for which no comparable data yet exists in HD mice or humans. These additional factors expand the potential druggable space for diseases like HD where somatic expansion is a significant contributor to disease impact.
- Published
- 2021
- Full Text
- View/download PDF
65. Repeat expansions confer WRN dependence in microsatellite-unstable cancers.
- Author
-
van Wietmarschen N, Sridharan S, Nathan WJ, Tubbs A, Chan EM, Callen E, Wu W, Belinky F, Tripathi V, Wong N, Foster K, Noorbakhsh J, Garimella K, Cruz-Migoni A, Sommers JA, Huang Y, Borah AA, Smith JT, Kalfon J, Kesten N, Fugger K, Walker RL, Dolzhenko E, Eberle MA, Hayward BE, Usdin K, Freudenreich CH, Brosh RM Jr, West SC, McHugh PJ, Meltzer PS, Bass AJ, and Nussenzweig A
- Subjects
- Ataxia Telangiectasia Mutated Proteins metabolism, Cell Line, Tumor, Chromosomes, Human genetics, Chromosomes, Human metabolism, Chromothripsis, DNA Cleavage, DNA Replication, DNA-Binding Proteins metabolism, Endodeoxyribonucleases metabolism, Endonucleases metabolism, Genomic Instability, Humans, Recombinases metabolism, DNA Breaks, Double-Stranded, DNA Repeat Expansion genetics, Dinucleotide Repeats genetics, Neoplasms genetics, Werner Syndrome Helicase metabolism
- Abstract
The RecQ DNA helicase WRN is a synthetic lethal target for cancer cells with microsatellite instability (MSI), a form of genetic hypermutability that arises from impaired mismatch repair
1-4 . Depletion of WRN induces widespread DNA double-strand breaks in MSI cells, leading to cell cycle arrest and/or apoptosis. However, the mechanism by which WRN protects MSI-associated cancers from double-strand breaks remains unclear. Here we show that TA-dinucleotide repeats are highly unstable in MSI cells and undergo large-scale expansions, distinct from previously described insertion or deletion mutations of a few nucleotides5 . Expanded TA repeats form non-B DNA secondary structures that stall replication forks, activate the ATR checkpoint kinase, and require unwinding by the WRN helicase. In the absence of WRN, the expanded TA-dinucleotide repeats are susceptible to cleavage by the MUS81 nuclease, leading to massive chromosome shattering. These findings identify a distinct biomarker that underlies the synthetic lethal dependence on WRN, and support the development of therapeutic agents that target WRN for MSI-associated cancers.- Published
- 2020
- Full Text
- View/download PDF
66. A point mutation in the nuclease domain of MLH3 eliminates repeat expansions in a mouse stem cell model of the Fragile X-related disorders.
- Author
-
Hayward BE, Steinbach PJ, and Usdin K
- Subjects
- Alleles, Animals, Cell Line, Disease Models, Animal, Male, Mice, Point Mutation, Protein Domains genetics, Stem Cells, Fragile X Syndrome genetics, MutL Proteins genetics, Trinucleotide Repeat Expansion
- Abstract
The Fragile X-related disorders (FXDs) are Repeat Expansion Diseases, genetic disorders that result from the expansion of a disease-specific microsatellite. In those Repeat Expansion Disease models where it has been examined, expansion is dependent on functional mismatch repair (MMR) factors, including MutLγ, a heterodimer of MLH1/MLH3, one of the three MutL complexes found in mammals and a minor player in MMR. In contrast, MutLα, a much more abundant MutL complex that is the major contributor to MMR, is either not required for expansion or plays a limited role in expansion in many model systems. How MutLγ acts to generate expansions is unclear given its normal role in protecting against microsatellite instability and while MLH3 does have an associated endonuclease activity, whether that contributes to repeat expansion is uncertain. We show here, using a gene-editing approach, that a point mutation that eliminates the endonuclease activity of MLH3 eliminates expansions in an FXD mouse embryonic stem cell model. This restricts the number of possible models for repeat expansion and supports the idea that MutLγ may be a useful druggable target to reduce somatic expansion in those disorders where it contributes to disease pathology., (Published by Oxford University Press on behalf of Nucleic Acids Research 2020.)
- Published
- 2020
- Full Text
- View/download PDF
67. CGG Repeat Expansion, and Elevated Fmr1 Transcription and Mitochondrial Copy Number in a New Fragile X PM Mouse Embryonic Stem Cell Model.
- Author
-
Gazy I, Miller CJ, Kim GY, and Usdin K
- Abstract
The Fragile-X related disorders (FXDs) are Repeat Expansion Diseases (REDs) that result from expansion of a CGG-repeat tract located at the 5' end of the FMR1 gene. While expansion affects transmission risk and can also affect disease risk and severity, the underlying molecular mechanism responsible is unknown. Despite the fact that expanded alleles can be seen both in humans and mouse models in vivo , existing patient-derived cells do not show significant repeat expansions even after extended periods in culture. In order to develop a good tissue culture model for studying expansions we tested whether mouse embryonic stem cells (mESCs) carrying an expanded CGG repeat tract in the endogenous Fmr1 gene are permissive for expansion. We show here that these mESCs have a very high frequency of expansion that allows changes in the repeat number to be seen within a matter of days. CRISPR-Cas9 gene editing of these cells suggests that this may be due in part to the fact that non-homologous end-joining (NHEJ), which is able to protect against expansions in some cell types, is not effective in mESCs. CRISPR-Cas9 gene editing also shows that these expansions are MSH2-dependent, consistent with those seen in vivo . While comparable human Genome Wide Association (GWA) studies are not available for the FXDs, such studies have implicated MSH2 in expansion in other REDs. The shared unusual requirement for MSH2 for this type of microsatellite instability suggests that this new cell-based system is relevant for understanding the mechanism responsible for this peculiar type of mutation in humans. The high frequency of expansions and the ease of gene editing these cells should expedite the identification of factors that affect expansion risk. Additionally, we found that, as with cells from human premutation (PM) carriers, these cell lines have elevated mitochondrial copy numbers and Fmr1 hyperexpression, that we show here is O
2 -sensitive. Thus, this new stem cell model should facilitate studies of both repeat expansion and the consequences of expansion during early embryonic development., (Copyright © 2020 Gazy, Miller, Kim and Usdin.)- Published
- 2020
- Full Text
- View/download PDF
68. All three mammalian MutL complexes are required for repeat expansion in a mouse cell model of the Fragile X-related disorders.
- Author
-
Miller CJ, Kim GY, Zhao X, and Usdin K
- Subjects
- Animals, Cell Line, DNA Mismatch Repair, Disease Models, Animal, Embryonic Stem Cells, Gene Knockout Techniques, Humans, Mice, Mismatch Repair Endonuclease PMS2 metabolism, MutL Proteins metabolism, Fragile X Syndrome genetics, Mismatch Repair Endonuclease PMS2 genetics, MutL Proteins genetics, Trinucleotide Repeat Expansion genetics
- Abstract
Expansion of a CGG-repeat tract in the 5' untranslated region of the FMR1 gene causes the fragile X-related disorders (FXDs; aka the FMR1 disorders). The expansion mechanism is likely shared by the 35+ other diseases resulting from expansion of a disease-specific microsatellite, but many steps in this process are unknown. We have shown previously that expansion is dependent upon functional mismatch repair proteins, including an absolute requirement for MutLγ, one of the three MutL heterodimeric complexes found in mammalian cells. We demonstrate here that both MutLα and MutLβ, the two other MutL complexes present in mammalian cells, are also required for most, if not all, expansions in a mouse embryonic stem cell model of the FXDs. A role for MutLα and MutLβ is consistent with human GWA studies implicating these complexes as modifiers of expansion risk in other Repeat Expansion Diseases. The requirement for all three complexes suggests a novel model in which these complexes co-operate to generate expansions. It also suggests that the PMS1 subunit of MutLβ may be a reasonable therapeutic target in those diseases in which somatic expansion is an important disease modifier., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
69. Molecular analysis of FMR1 alleles for fragile X syndrome diagnosis and patient stratification.
- Author
-
Kumari D and Usdin K
- Subjects
- Female, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome epidemiology, Fragile X Syndrome metabolism, Gene Expression Regulation, Genetic Association Studies, Genetic Predisposition to Disease, Genetic Testing, Humans, Male, RNA, Messenger genetics, Alleles, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Mutation
- Published
- 2020
- Full Text
- View/download PDF
70. Small Molecules Targeting H3K9 Methylation Prevent Silencing of Reactivated FMR1 Alleles in Fragile X Syndrome Patient Derived Cells.
- Author
-
Kumari D, Sciascia N, and Usdin K
- Subjects
- Alleles, Cells, Cultured, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome drug therapy, Fragile X Syndrome pathology, Humans, Trinucleotide Repeats, DNA Methylation, Fragile X Mental Retardation Protein antagonists & inhibitors, Fragile X Syndrome genetics, Gene Silencing, Pharmaceutical Preparations metabolism, Small Molecule Libraries pharmacology
- Abstract
In fragile X syndrome (FXS), expansion of a CGG repeat tract in the 5'-untranslated region of the FMR1 gene to >200 repeats causes transcriptional silencing by inducing heterochromatin formation. Understanding the mechanism of FMR1 silencing is important as gene reactivation is a potential treatment approach for FXS. To date, only the DNA demethylating drug 5-azadeoxycytidine (AZA) has proved effective at gene reactivation; however, this drug is toxic. The repressive H3K9 methylation mark is enriched on the FMR1 gene in FXS patient cells and is thus a potential druggable target. However, its contribution to the silencing process is unclear. Here, we studied the effect of small molecule inhibitors of H3K9 methylation on FMR1 expression in FXS patient cells. Chaetocin showed a small effect on FMR1 gene reactivation and a synergistic effect on FMR1 mRNA levels when used in combination with AZA. Additionally, chaetocin, BIX01294 and 3-Deazaneplanocin A (DZNep) were able to significantly delay the re-silencing of AZA-reactivated FMR1 alleles. These data are consistent with the idea that H3K9 methylation precedes DNA methylation and that removal of DNA methylation is necessary to see the optimal effect of histone methyl-transferase (HMT) inhibitors on FMR1 gene expression. Nonetheless, our data also show that drugs targeting repressive H3K9 methylation marks are able to produce sustained reactivation of the FMR1 gene after a single dose of AZA.
- Published
- 2020
- Full Text
- View/download PDF
71. Isolation and Analysis of the CGG-Repeat Size in Male and Female Gametes from a Fragile X Mouse Model.
- Author
-
Zhao X, Lu H, Dagur PK, and Usdin K
- Subjects
- Animals, Cell Separation, Disease Models, Animal, Female, Flow Cytometry, Humans, Male, Mice, Oocytes chemistry, Single-Cell Analysis, Spermatocytes chemistry, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Oocytes cytology, Spermatocytes cytology, Trinucleotide Repeat Expansion
- Abstract
Analysis of individual gametes has a number of applications in the study of the mechanism of repeat expansion in mouse models of the fragile X-related disorders, as well as in mouse models of other repeat expansion diseases. This chapter describes the techniques required to isolate oocytes and male gametes of different stages of maturity, along with the techniques required to accurately determine the repeat number in these gametes.
- Published
- 2020
- Full Text
- View/download PDF
72. Fragile X syndrome in a male with methylated premutation alleles and no detectable methylated full mutation alleles.
- Author
-
Hayward B, Loutaev I, Ding X, Nolin SL, Thurm A, Usdin K, and Smith CB
- Subjects
- Child, Preschool, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome psychology, Humans, Male, Young Adult, Alleles, DNA Methylation genetics, Fragile X Syndrome genetics, Mutation genetics
- Abstract
Most cases of fragile X syndrome (FXS) result from aberrant methylation of the FMR1 gene. Methylation occurs when the number of tandemly arranged cytosine guanine guanine (CGG)-repeats in the 5' end of the transcriptional unit of FMR1 exceeds a certain critical threshold, thought to be between 200 and 400 repeats. Such alleles are referred to as full mutation (FM) alleles. Premutation (PM) alleles, alleles with 55-200 repeats, are generally not aberrantly methylated and in fact may have hyperexpression of the FMR1 mRNA. We describe here a male who meets the diagnostic criteria for FXS, who is highly mosaic with a mixture of multiple PM and FM alleles and 50% methylation. However, the methylated alleles are limited to two alleles in the PM range, ~165 and ~175 repeats respectively, with the FM alleles being unmethylated. This finding has implications for FXS diagnosis as well as for efforts to delete the repeat in individuals with FXS using a CRISPR-Cas9 approach., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
73. Glutaminase Deficiency Caused by Short Tandem Repeat Expansion in GLS. Reply.
- Author
-
van Kuilenburg ABP, Usdin K, and van Karnebeek CDM
- Subjects
- Glutamine, Glutaminase, Microsatellite Repeats
- Published
- 2019
- Full Text
- View/download PDF
74. Repeat Instability in the Fragile X-Related Disorders: Lessons from a Mouse Model.
- Author
-
Zhao X, Gazy I, Hayward B, Pintado E, Hwang YH, Tassone F, and Usdin K
- Abstract
The fragile X-related disorders (FXDs) are a group of clinical conditions that result primarily from an unusual mutation, the expansion of a CGG-repeat tract in exon 1 of the FMR1 gene. Mouse models are proving useful for understanding many aspects of disease pathology in these disorders. There is also reason to think that such models may be useful for understanding the molecular basis of the unusual mutation responsible for these disorders. This review will discuss what has been learnt to date about mechanisms of repeat instability from a knock-in FXD mouse model and what the implications of these findings may be for humans carrying expansion-prone FMR1 alleles.
- Published
- 2019
- Full Text
- View/download PDF
75. Pharmacological Reactivation of the Silenced FMR1 Gene as a Targeted Therapeutic Approach for Fragile X Syndrome.
- Author
-
Kumari D, Gazy I, and Usdin K
- Abstract
More than ~200 CGG repeats in the 5' untranslated region of the FMR1 gene results in transcriptional silencing and the absence of the FMR1 encoded protein, FMRP. FMRP is an RNA-binding protein that regulates the transport and translation of a variety of brain mRNAs in an activity-dependent manner. The loss of FMRP causes dysregulation of many neuronal pathways and results in an intellectual disability disorder, fragile X syndrome (FXS). Currently, there is no effective treatment for FXS. In this review, we discuss reactivation of the FMR1 gene as a potential approach for FXS treatment with an emphasis on the use of small molecules to inhibit the pathways important for gene silencing.
- Published
- 2019
- Full Text
- View/download PDF
76. Double-strand break repair plays a role in repeat instability in a fragile X mouse model.
- Author
-
Gazy I, Hayward B, Potapova S, Zhao X, and Usdin K
- Subjects
- Animals, DNA End-Joining Repair, DNA Ligase ATP metabolism, Disease Models, Animal, Hepatocytes metabolism, Homologous Recombination, Male, Mice, Mice, Inbred C57BL, DNA Breaks, Double-Stranded, DNA Repair, Fragile X Syndrome genetics, Repetitive Sequences, Nucleic Acid
- Abstract
Expansion of a CGG-repeat tract in the 5' UTR of FMR1 is responsible for the Fragile X-related disorders (FXDs), FXTAS, FXPOI and FXS. Previous work in a mouse model of these disorders has implicated proteins in the base excision and the mismatch repair (MMR) pathways in the expansion mechanism. However, the precise role of these factors in this process is not well understood. The essential role of MutLγ, a complex that plays a minor role in MMR but that is essential for resolving Holliday junctions during meiosis, raises the possibility that expansions proceed via a Holliday junction-like intermediate that is processed to generate a double-strand break (DSB). We show here in an FXD mouse model that LIG4, a ligase essential for non-homologous end-joining (NHEJ), a form of DSB repair (DSBR), protects against expansions. However, a mutation in MRE11, a nuclease that is important for several other DSBR pathways including homologous recombination (HR), has no effect on the extent of expansion. Our results suggest that the expansion pathway competes with NHEJ for the processing of a DSB intermediate. Thus, expansion likely proceeds via an NHEJ-independent DSBR pathway that may also be HR-independent., (Published by Elsevier B.V.)
- Published
- 2019
- Full Text
- View/download PDF
77. Assays for Determining Repeat Number, Methylation Status, and AGG Interruptions in the Fragile X-Related Disorders.
- Author
-
Hayward BE and Usdin K
- Subjects
- Humans, DNA Methylation, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Trinucleotide Repeat Expansion
- Abstract
Knowledge of the CGG•CCG-repeat number, AGG interruption status, and the extent of DNA methylation of the FMR1 gene are vital for both diagnosis of the fragile X-related disorders and for basic research into disease mechanisms. We describe here assays that we use in our laboratory to assess these parameters. Our assays are PCR-based and include one for repeat size that can also be used to assess the extent of methylation and a related assay that allows the AGG interruption pattern to be reliably determined even in women. A second more quantitative methylation assay is also described. We also describe our method for cloning of repeats to generate the reference standards necessary for the accurate determination of repeat number and AGG interruption status.
- Published
- 2019
- Full Text
- View/download PDF
78. MutLγ promotes repeat expansion in a Fragile X mouse model while EXO1 is protective.
- Author
-
Zhao X, Zhang Y, Wilkins K, Edelmann W, and Usdin K
- Subjects
- Animals, DNA Mismatch Repair genetics, DNA Mismatch Repair physiology, DNA Repair, DNA Repair Enzymes physiology, Disease Models, Animal, Exodeoxyribonucleases physiology, Fragile X Mental Retardation Protein genetics, Genomic Instability, Mice, Mice, Inbred C57BL, Mice, Knockout, MutL Protein Homolog 1 metabolism, MutL Proteins metabolism, Mutation, Trinucleotide Repeat Expansion genetics, DNA Repair Enzymes genetics, Exodeoxyribonucleases genetics, Fragile X Syndrome genetics, MutL Proteins genetics
- Abstract
The Fragile X-related disorders (FXDs) are Repeat Expansion Diseases resulting from an expansion of a CGG-repeat tract at the 5' end of the FMR1 gene. The mechanism responsible for this unusual mutation is not fully understood. We have previously shown that mismatch repair (MMR) complexes, MSH2/MSH3 (MutSβ) and MSH2/MSH6 (MutSα), together with Polβ, a DNA polymerase important for base excision repair (BER), are important for expansions in a mouse model of these disorders. Here we show that MLH1/MLH3 (MutLγ), a protein complex that can act downstream of MutSβ in MMR, is also required for all germ line and somatic expansions. However, exonuclease I (EXO1), which acts downstream of MutL proteins in MMR, is not required. In fact, a null mutation in Exo1 results in more extensive germ line and somatic expansions than is seen in Exo1+/+ animals. Furthermore, mice homozygous for a point mutation (D173A) in Exo1 that eliminates its nuclease activity but retains its native conformation, shows a level of expansion that is intermediate between Exo1+/+ and Exo1-/- animals. Thus, our data suggests that expansion of the FX repeat in this mouse model occurs via a MutLγ-dependent, EXO1-independent pathway, with EXO1 protecting against expansion both in a nuclease-dependent and a nuclease-independent manner. Our data thus have implications for the expansion mechanism and add to our understanding of the genetic factors that may be modifiers of expansion risk in humans., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
79. FAN1 protects against repeat expansions in a Fragile X mouse model.
- Author
-
Zhao XN and Usdin K
- Subjects
- Animals, DNA metabolism, Disease Models, Animal, Endodeoxyribonucleases genetics, Exodeoxyribonucleases, Female, Fragile X Syndrome genetics, Male, Mice, Mice, Knockout, Multifunctional Enzymes, Mutation, DNA Mismatch Repair, Endodeoxyribonucleases metabolism, Fragile X Syndrome metabolism, Trinucleotide Repeat Expansion
- Abstract
The Fragile X-related disorders (FXDs) are members of a large group of human neurological or neurodevelopmental conditions known as the Repeat Expansion Diseases. The mutation responsible for all of these diseases is an expansion in the size of a disease-specific tandem repeat tract. However, the underlying cause of this unusual mutation is unknown. Genome-wide association studies have identified single nucleotide polymorphisms (SNPs) in the vicinity of the FAN1 (MIM* 613534) gene that are associated with variations in the age at onset of a number of Repeat Expansion Diseases. FAN1 is a nuclease that has both 5'-3' exonuclease and 5' flap endonuclease activities. Here we show in a model for the FXDs that Fan1
-/- mice have expansions that, in some tissues including brain, are 2-3 times as extensive as they are in Fan1+/+ mice. However, no effect of the loss of FAN1 was apparent for germ line expansions. Thus, FAN1 plays an important role in protecting against somatic expansions but is either not involved in protecting against intergenerational repeat expansions or is redundant with other related enzymes. However, since loss of FAN1 results in increased expansions in brain and other somatic tissue, FAN1 polymorphisms may be important disease modifiers in those Repeat Expansion Diseases in which somatic expansion contributes to age at onset or disease severity., (Published by Elsevier B.V.)- Published
- 2018
- Full Text
- View/download PDF
80. Timing of Expansion of Fragile X Premutation Alleles During Intergenerational Transmission in a Mouse Model of the Fragile X-Related Disorders.
- Author
-
Zhao XN and Usdin K
- Abstract
Fragile X syndrome (FXS) is caused by the maternal expansion of an unstable CGG-repeat tract located in the first exon of the FMR1 gene. Further changes in repeat number occur during embryogenesis resulting in individuals sometimes being highly mosaic. Here we show in a mouse model that, in males, expansions are already present in primary spermatocytes with no additional expansions occurring in later stages of gametogenesis. We also show that, in females, expansion occurs in the post-natal oocyte. Additional expansions and a high frequency of large contractions are seen in two-cell stage embryos. Expansion in oocytes, which are non-dividing, would be consistent with a mechanism involving aberrant DNA repair or recombination rather than a problem with chromosomal replication. Given the difficulty of replicating large CGG-repeat tracts, we speculate that very large expanded alleles may be prone to contract in the mitotically proliferating spermatagonial stem cells in men. However, expanded alleles may not be under such pressure in the non-dividing oocyte. The high degree of both expansions and contractions seen in early embryos may contribute to the high frequency of somatic mosaicism that is observed in humans. Our data thus suggest an explanation for the fact that FXS is exclusively maternally transmitted and lend support to models for repeat expansion that are based on problems arising during DNA repair.
- Published
- 2018
- Full Text
- View/download PDF
81. Improved Assays for AGG Interruptions in Fragile X Premutation Carriers.
- Author
-
Hayward BE and Usdin K
- Subjects
- Alleles, Female, Fragile X Syndrome genetics, Fragile X Syndrome pathology, Heterozygote, Humans, Male, Mosaicism, Mutation, Polymerase Chain Reaction methods, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Genetic Counseling, Trinucleotide Repeat Expansion genetics
- Abstract
The learning disability fragile X syndrome results from the presence of >200 CGG/CCG repeats in exon 1 of the X-linked gene FMR1. Such alleles arise by expansion from maternally transmitted FMR1 premutation alleles, alleles having 55 to 200 repeats. Expansion risk is directly related to maternal repeat number. However, AGG interruptions to the repeat tract are important modifiers of expansion risk. Thus, the ability to identify such interruptions is crucial for the appropriate genetic counseling of females who are premutation carriers. First-generation triplet-primed PCR assays allow these interruptions to be detected. However, because the triplet primer used has multiple binding sites in the repeat tract, interpreting the results is not straightforward and it is not always possible to unambiguously determine the AGG-interruption status in females because of the difficulties associated with the presence of a second X chromosome. Interpretation is further complicated by any repeat size mosaicism that may be present. We have developed second-generation PCR assays that prime specifically at the interruptions. These assays are simpler to interpret and better able to evaluate this important determinant of expansion risk in females even in those with a mixture of premutation allele sizes., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
82. Recent advances in assays for the fragile X-related disorders.
- Author
-
Hayward BE, Kumari D, and Usdin K
- Subjects
- Female, Humans, Male, Ataxia genetics, Ataxia metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Gene Silencing, Primary Ovarian Insufficiency genetics, Primary Ovarian Insufficiency metabolism, RNA Stability, Tremor genetics, Tremor metabolism
- Abstract
The fragile X-related disorders are a group of three clinical conditions resulting from the instability of a CGG-repeat tract at the 5' end of the FMR1 transcript. Fragile X-associated tremor/ataxia syndrome (FXTAS) and fragile X-associated primary ovarian insufficiency (FXPOI) are disorders seen in carriers of FMR1 alleles with 55-200 repeats. Female carriers of these premutation (PM) alleles are also at risk of having a child who has an FMR1 allele with >200 repeats. Most of these full mutation (FM) alleles are epigenetically silenced resulting in a deficit of the FMR1 gene product, FMRP. This results in fragile X Syndrome (FXS), the most common heritable cause of intellectual disability and autism. The diagnosis and study of these disorders is challenging, in part because the detection of alleles with large repeat numbers has, until recently, been either time-consuming or unreliable. This problem is compounded by the mosaicism for repeat length and/or DNA methylation that is frequently seen in PM and FM carriers. Furthermore, since AGG interruptions in the repeat tract affect the risk that a FM allele will be maternally transmitted, the ability to accurately detect these interruptions in female PM carriers is an additional challenge that must be met. This review will discuss some of the pros and cons of some recently described assays for these disorders, including those that detect FMRP levels directly, as well as emerging technologies that promise to improve the diagnosis of these conditions and to be useful in both basic and translational research settings.
- Published
- 2017
- Full Text
- View/download PDF
83. CGG-repeat dynamics and FMR1 gene silencing in fragile X syndrome stem cells and stem cell-derived neurons.
- Author
-
Zhou Y, Kumari D, Sciascia N, and Usdin K
- Subjects
- 5' Untranslated Regions, Alleles, Cell Differentiation, Cell Line, DNA Methylation, Embryonic Stem Cells pathology, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome metabolism, Fragile X Syndrome pathology, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Male, Neurons pathology, Primary Cell Culture, Time Factors, Embryonic Stem Cells metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Gene Silencing, Neurons metabolism, Trinucleotide Repeat Expansion
- Abstract
Background: Fragile X syndrome (FXS), a common cause of intellectual disability and autism, results from the expansion of a CGG-repeat tract in the 5' untranslated region of the FMR1 gene to >200 repeats. Such expanded alleles, known as full mutation (FM) alleles, are epigenetically silenced in differentiated cells thus resulting in the loss of FMRP, a protein important for learning and memory. The timing of repeat expansion and FMR1 gene silencing is controversial., Methods: We monitored the repeat size and methylation status of FMR1 alleles with expanded CGG repeats in patient-derived induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) that were grown for extended period of time either as stem cells or differentiated into neurons. We used a PCR assay optimized for the amplification of large CGG repeats for sizing, and a quantitative methylation-specific PCR for the analysis of FMR1 promoter methylation. The FMR1 mRNA levels were analyzed by qRT-PCR. FMRP levels were determined by western blotting and immunofluorescence. Chromatin immunoprecipitation was used to study the association of repressive histone marks with the FMR1 gene in FXS ESCs., Results: We show here that while FMR1 gene silencing can be seen in FXS embryonic stem cells (ESCs), some silenced alleles contract and when the repeat number drops below ~400, DNA methylation erodes, even when the repeat number remains >200. The resultant active alleles do not show the large step-wise expansions seen in stem cells from other repeat expansion diseases. Furthermore, there may be selection against large active alleles and these alleles do not expand further or become silenced on neuronal differentiation., Conclusions: Our data support the hypotheses that (i) large expansions occur prezygotically or in the very early embryo, (ii) large unmethylated alleles may be deleterious in stem cells, (iii) methylation can occur on alleles with >400 repeats very early in embryogenesis, and (iv) expansion and contraction may occur by different mechanisms. Our data also suggest that the threshold for stable methylation of FM alleles may be higher than previously thought. A higher threshold might explain why some carriers of FM alleles escape methylation. It may also provide a simple explanation for why silencing has not been observed in mouse models with >200 repeats.
- Published
- 2016
- Full Text
- View/download PDF
84. Ups and Downs: Mechanisms of Repeat Instability in the Fragile X-Related Disorders.
- Author
-
Zhao XN and Usdin K
- Abstract
The Fragile X-related disorders (FXDs) are a group of clinical conditions resulting from the expansion of a CGG/CCG-repeat tract in exon 1 of the Fragile X mental retardation 1 (FMR1) gene. While expansions of the repeat tract predominate, contractions are also seen with the net result being that individuals can show extensive repeat length heterogeneity in different tissues. The mechanisms responsible for expansion and contraction are still not well understood. This review will discuss what is known about these processes and current evidence that supports a model in which expansion arises from the interaction of components of the base excision repair, mismatch repair and transcription coupled repair pathways., Competing Interests: The authors have no conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
85. A Set of Assays for the Comprehensive Analysis of FMR1 Alleles in the Fragile X-Related Disorders.
- Author
-
Hayward BE, Zhou Y, Kumari D, and Usdin K
- Subjects
- Cell Line, DNA Methylation, Female, Humans, Male, Polymerase Chain Reaction methods, Promoter Regions, Genetic, Trinucleotide Repeat Expansion, Trinucleotide Repeats, Workflow, Alleles, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Genetic Testing methods, Mutation
- Abstract
The diagnosis and study of the fragile X-related disorders is complicated by the difficulty of amplifying the long CGG/CCG-repeat tracts that are responsible for disease pathology, the potential presence of AGG interruptions within the repeat tract that can ameliorate expansion risk, the occurrence of variable DNA methylation that modulates disease severity, and the high frequency of mosaicism for both repeat number and methylation status. These factors complicate patient risk assessment. In addition, the variability in these parameters that is seen when patient cells are grown in culture requires their frequent monitoring to ensure reproducible results in a research setting. Many existing assays have the limited ability to amplify long alleles, particularly in a mixture of different allele sizes. Others are better at this, but are too expensive for routine use in most laboratories or for newborn screening programs and use reagents that are proprietary. We describe herein a set of assays to routinely evaluate all of these important parameters in a time- and cost-effective way., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
86. Sustained expression of FMR1 mRNA from reactivated fragile X syndrome alleles after treatment with small molecules that prevent trimethylation of H3K27.
- Author
-
Kumari D and Usdin K
- Subjects
- Azacitidine pharmacology, Cell Line, Tumor, DNA Methylation, Decitabine, Gene Expression Regulation drug effects, Humans, Male, Methylation drug effects, Microsatellite Repeats drug effects, Up-Regulation, Azacitidine analogs & derivatives, Enhancer of Zeste Homolog 2 Protein metabolism, Fragile X Mental Retardation Protein genetics, Histones metabolism, Small Molecule Libraries pharmacology
- Abstract
Expansion of a CGG-repeat tract in the 5'-untranslated region of the FMR1 gene to >200 repeats results in epigenetic silencing of the gene by a mechanism that is still unknown. FMR1 gene silencing results in fragile X syndrome (FXS), the most common heritable cause of intellectual disability. We have previously shown that reactivation of the FMR1 gene in FXS cells with 5-azadeoxycytidine (AZA) leads to the transient recruitment of EZH2, the polycomb repressive complex 2 (PRC2) component responsible for H3K27 trimethylation, and that this recruitment depends on the presence of the FMR1 transcript. However, whether H3K27 trimethylation was essential for FMR1 re-silencing was not known. We show here that EZH2 inhibitors increased FMR1 expression and significantly delayed re-silencing of the FMR1 gene in AZA-treated FXS cells. This delay occurred despite the fact that EZH2 inhibition did not prevent the return of DNA methylation. Treatment with compound 1a, a small molecule that targets CGG-repeats in the FMR1 mRNA, also resulted in sustained expression of the FMR1 gene in AZA-treated cells. This effect of 1a was also associated with a decrease in the levels of H3K27 trimethylation but not DNA methylation. Thus, our data show that EZH2 plays a critical role in the FMR1 gene silencing process and that its inhibition can prolong expression of the FMR1 gene even in the presence of its transcript., (© The Author 2016. Published by Oxford University Press. This work is written by US Government employees and is in the public domain in the United States.)
- Published
- 2016
- Full Text
- View/download PDF
87. A MutSβ-Dependent Contribution of MutSα to Repeat Expansions in Fragile X Premutation Mice?
- Author
-
Zhao XN, Lokanga R, Allette K, Gazy I, Wu D, and Usdin K
- Subjects
- Animals, Disease Models, Animal, Female, Gene Expression Regulation, Genotype, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, MutS Homolog 2 Protein genetics, MutS Homolog 3 Protein, Mutation, Oligonucleotides genetics, Proteins genetics, DNA-Binding Proteins genetics, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, MutS DNA Mismatch-Binding Protein genetics
- Abstract
The fragile X-related disorders result from expansion of a CGG/CCG microsatellite in the 5' UTR of the FMR1 gene. We have previously demonstrated that the MSH2/MSH3 complex, MutSβ, that is important for mismatch repair, is essential for almost all expansions in a mouse model of these disorders. Here we show that the MSH2/MSH6 complex, MutSα also contributes to the production of both germ line and somatic expansions as evidenced by the reduction in the number of expansions observed in Msh6-/- mice. This effect is not mediated via an indirect effect of the loss of MSH6 on the level of MSH3. However, since MutSβ is required for 98% of germ line expansions and almost all somatic ones, MutSα is apparently not able to efficiently substitute for MutSβ in the expansion process. Using purified human proteins we demonstrate that MutSα, like MutSβ, binds to substrates with loop-outs of the repeats and increases the thermal stability of the structures that they form. We also show that MutSα facilitates binding of MutSβ to these loop-outs. These data suggest possible models for the contribution of MutSα to repeat expansion. In addition, we show that unlike MutSβ, MutSα may also act to protect against repeat contractions in the Fmr1 gene.
- Published
- 2016
- Full Text
- View/download PDF
88. Granulosa cell and oocyte mitochondrial abnormalities in a mouse model of fragile X primary ovarian insufficiency.
- Author
-
Conca Dioguardi C, Uslu B, Haynes M, Kurus M, Gul M, Miao DQ, De Santis L, Ferrari M, Bellone S, Santin A, Giulivi C, Hoffman G, Usdin K, and Johnson J
- Subjects
- Animals, Disease Models, Animal, Female, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, Fragile X Syndrome pathology, Granulosa Cells pathology, Mice, Mice, Mutant Strains, Mitochondria pathology, Oocytes pathology, Ovarian Follicle cytology, Ovarian Follicle metabolism, Ovary metabolism, Ovary pathology, Reverse Transcriptase Polymerase Chain Reaction, Granulosa Cells metabolism, Mitochondria metabolism, Oocytes metabolism, Primary Ovarian Insufficiency metabolism, Primary Ovarian Insufficiency pathology
- Abstract
Study Hypothesis: We hypothesized that the mitochondria of granulosa cells (GC) and/or oocytes might be abnormal in a mouse model of fragile X premutation (FXPM)., Study Finding: Mice heterozygous and homozygous for the FXPM have increased death (atresia) of large ovarian follicles, fewer corpora lutea with a gene dosage effect manifesting in decreased litter size(s). Furthermore, granulosa cells (GC) and oocytes of FXPM mice have decreased mitochondrial content, structurally abnormal mitochondria, and reduced expression of critical mitochondrial genes. Because this mouse allele produces the mutant Fragile X mental retardation 1 (Fmr1) transcript and reduced levels of wild-type (WT) Fmr1 protein (FMRP), but does not produce a Repeat Associated Non-ATG Translation (RAN)-translation product, our data lend support to the idea that Fmr1 mRNA with large numbers of CGG-repeats is intrinsically deleterious in the ovary., What Is Known Already: Mitochondrial dysfunction has been detected in somatic cells of human and mouse FX PM carriers and mitochondria are essential for oogenesis and ovarian follicle development, FX-associated primary ovarian insufficiency (FXPOI) is seen in women with FXPM alleles. These alleles have 55-200 CGG repeats in the 5' UTR of an X-linked gene known as FMR1. The molecular basis of the pathology seen in this disorder is unclear but is thought to involve either some deleterious consequence of overexpression of RNA with long CGG-repeat tracts or of the generation of a repeat-associated non-AUG translation (RAN translation) product that is toxic., Study Design, Samples/materials, Methods: Analysis of ovarian function in a knock-in FXPM mouse model carrying 130 CGG repeats was performed as follows on WT, PM/+, and PM/PM genotypes. Histomorphometric assessment of follicle and corpora lutea numbers in ovaries from 8-month-old mice was executed, along with litter size analysis. Mitochondrial DNA copy number was quantified in oocytes and GC using quantitative PCR, and cumulus granulosa mitochondrial content was measured by flow cytometric analysis after staining of cells with Mitotracker dye. Transmission electron micrographs were prepared of GC within small growing follicles and mitochondrial architecture was compared. Quantitative RT-PCR analysis of key genes involved in mitochondrial structure and recycling was performed., Main Results and the Role of Chance: A defect was found in follicle survival at the large antral stage in PM/+ and PM/PM mice. Litter size was significantly decreased in PM/PM mice, and corpora lutea were significantly reduced in mice of both mutant genotypes. Mitochondrial DNA copy number was significantly decreased in GC and metaphase II eggs in mutants. Flow cytometric analysis revealed that PM/+ and PM/PM animals lack the cumulus GC that harbor the greatest mitochondrial content as found in wild-type animals. Electron microscopic evaluation of GC of small growing follicles revealed mitochondrial structural abnormalities, including disorganized and vacuolar cristae. Finally, aberrant mitochondrial gene expression was detected. Mitofusin 2 (Mfn2) and Optic atrophy 1 (Opa1), genes involved in mitochondrial fusion and structure, respectively, were significantly decreased in whole ovaries of both mutant genotypes. Mitochondrial fission factor 1 (Mff1) was significantly decreased in PM/+ and PM/PM GC and eggs compared with wild-type controls., Limitations, Reasons for Caution: Data from the mouse model used for these studies should be viewed with some caution when considering parallels to the human FXPOI condition., Wider Implications of the Findings: Our data lend support to the idea that Fmr1 mRNA with large numbers of CGG-repeats is intrinsically deleterious in the ovary. FXPM disease states, including FXPOI, may share mitochondrial dysfunction as a common underlying mechanism., Large Scale Data: Not applicable., Study Funding and Competing Interests: Studies were supported by NIH R21 071873 (J.J./G.H), The Albert McKern Fund for Perinatal Research (J.J.), NIH Intramural Funds (K.U.), and a TUBITAK Research Fellowship Award (B.U.). No conflict(s) of interest or competing interest(s) are noted., (© The Author 2016. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
89. Mutsβ generates both expansions and contractions in a mouse model of the Fragile X-associated disorders.
- Author
-
Zhao XN, Kumari D, Gupta S, Wu D, Evanitsky M, Yang W, and Usdin K
- Subjects
- Animals, Cell Line, Chromosomal Instability, Disease Models, Animal, Female, Fragile X Syndrome physiopathology, Germ-Line Mutation, Male, Mice, Mice, Mutant Strains, MutS Homolog 3 Protein, Nucleic Acid Conformation, Protein Binding, Proteins genetics, Fragile X Syndrome genetics, MutS DNA Mismatch-Binding Protein physiology, Proteins physiology, Trinucleotide Repeat Expansion
- Abstract
Fragile X-associated disorders are Repeat Expansion Diseases that result from expansion of a CGG/CCG-repeat in the FMR1 gene. Contractions of the repeat tract also occur, albeit at lower frequency. However, these contractions can potentially modulate disease symptoms or generate an allele with repeat numbers in the normal range. Little is known about the expansion mechanism and even less about contractions. We have previously demonstrated that the mismatch repair (MMR) protein MSH2 is required for expansions in a mouse model of these disorders. Here, we show that MSH3, the MSH2-binding partner in the MutSβ complex, is required for 98% of germ line expansions and all somatic expansions in this model. In addition, we provide evidence for two different contraction mechanisms that operate in the mouse model, a MutSβ-independent one that generates small contractions and a MutSβ-dependent one that generates larger ones. We also show that MutSβ complexes formed with the repeats have altered kinetics of ATP hydrolysis relative to complexes with bona fide MMR substrates and that MutSβ increases the stability of the CCG-hairpins at physiological temperatures. These data may have important implications for our understanding of the mechanism(s) of repeat instability and for the role of MMR proteins in this process., (Published by Oxford University Press 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2015
- Full Text
- View/download PDF
90. Evidence for chromosome fragility at the frataxin locus in Friedreich ataxia.
- Author
-
Kumari D, Hayward B, Nakamura AJ, Bonner WM, and Usdin K
- Subjects
- Base Sequence, Cell Line, Humans, In Situ Hybridization, Fluorescence, Molecular Sequence Data, Sequence Analysis, DNA, Trinucleotide Repeat Expansion, Frataxin, Chromosome Fragility genetics, Chromosomes, Human, Pair 9 genetics, Friedreich Ataxia genetics, Iron-Binding Proteins genetics
- Abstract
Friedreich ataxia (FRDA) is a member of the Repeat Expansion Diseases, a group of genetic conditions resulting from an increase/expansion in the size of a specific tandem array. FRDA results from expansion of a GAA/TTC-tract in the first intron of the frataxin gene (FXN). The disease-associated tandem repeats all form secondary structures that are thought to contribute to the propensity of the repeat to expand. The subset of these diseases that result from a CGG/CCG-repeat expansion, such as Fragile X syndrome, also express a folate-sensitive fragile site coincident with the repeat on the affected chromosome. This chromosome fragility involves the generation of chromosome/chromatid gaps or breaks, or the high frequency loss of one or both copies of the affected gene when cells are grown under folate stress or as we showed previously, in the presence of an inhibitor of the ATM checkpoint kinase. Whether Repeat Expansion Disease loci containing different repeats form similar fragile sites was not known. We show here that the region of chromosome 9 that contains the FXN locus is intrinsically prone to breakage in vivo even in control cells. However, like FXS alleles, FRDA alleles show significantly elevated levels of chromosome abnormalities in the presence of an ATM inhibitor, consistent with the formation of a fragile site., (Published by Elsevier B.V.)
- Published
- 2015
- Full Text
- View/download PDF
91. The Repeat Expansion Diseases: The dark side of DNA repair.
- Author
-
Zhao XN and Usdin K
- Subjects
- Animals, Base Pair Mismatch, DNA Damage, Disease Models, Animal, Fragile X Syndrome pathology, Heredodegenerative Disorders, Nervous System pathology, Humans, Mice, Muscular Dystrophies pathology, Spinocerebellar Ataxias pathology, DNA Repair, Fragile X Syndrome genetics, Genome, Heredodegenerative Disorders, Nervous System genetics, Muscular Dystrophies genetics, Spinocerebellar Ataxias genetics, Trinucleotide Repeat Expansion
- Abstract
DNA repair normally protects the genome against mutations that threaten genome integrity and thus cell viability. However, growing evidence suggests that in the case of the Repeat Expansion Diseases, disorders that result from an increase in the size of a disease-specific microsatellite, the disease-causing mutation is actually the result of aberrant DNA repair. A variety of proteins from different DNA repair pathways have thus far been implicated in this process. This review will summarize recent findings from patients and from mouse models of these diseases that shed light on how these pathways may interact to cause repeat expansion., (Published by Elsevier B.V.)
- Published
- 2015
- Full Text
- View/download PDF
92. High-Throughput Screening to Identify Compounds That Increase Fragile X Mental Retardation Protein Expression in Neural Stem Cells Differentiated From Fragile X Syndrome Patient-Derived Induced Pluripotent Stem Cells.
- Author
-
Kumari D, Swaroop M, Southall N, Huang W, Zheng W, and Usdin K
- Abstract
Unlabelled: : Fragile X syndrome (FXS), the most common form of inherited cognitive disability, is caused by a deficiency of the fragile X mental retardation protein (FMRP). In most patients, the absence of FMRP is due to an aberrant transcriptional silencing of the fragile X mental retardation 1 (FMR1) gene. FXS has no cure, and the available treatments only provide symptomatic relief. Given that FMR1 gene silencing in FXS patient cells can be partially reversed by treatment with compounds that target repressive epigenetic marks, restoring FMRP expression could be one approach for the treatment of FXS. We describe a homogeneous and highly sensitive time-resolved fluorescence resonance energy transfer assay for FMRP detection in a 1,536-well plate format. Using neural stem cells differentiated from an FXS patient-derived induced pluripotent stem cell (iPSC) line that does not express any FMRP, we screened a collection of approximately 5,000 known tool compounds and approved drugs using this FMRP assay and identified 6 compounds that modestly increase FMR1 gene expression in FXS patient cells. Although none of these compounds resulted in clinically relevant levels of FMR1 mRNA, our data provide proof of principle that this assay combined with FXS patient-derived neural stem cells can be used in a high-throughput format to identify better lead compounds for FXS drug development., Significance: In this study, a specific and sensitive fluorescence resonance energy transfer-based assay for fragile X mental retardation protein detection was developed and optimized for high-throughput screening (HTS) of compound libraries using fragile X syndrome (FXS) patient-derived neural stem cells. The data suggest that this HTS format will be useful for the identification of better lead compounds for developing new therapeutics for FXS. This assay can also be adapted for FMRP detection in clinical and research settings., (©AlphaMed Press.)
- Published
- 2015
- Full Text
- View/download PDF
93. Repeat-mediated epigenetic dysregulation of the FMR1 gene in the fragile X-related disorders.
- Author
-
Usdin K and Kumari D
- Abstract
The fragile X-related disorders are members of the Repeat Expansion Diseases, a group of genetic conditions resulting from an expansion in the size of a tandem repeat tract at a specific genetic locus. The repeat responsible for disease pathology in the fragile X-related disorders is CGG/CCG and the repeat tract is located in the 5' UTR of the FMR1 gene, whose protein product FMRP, is important for the proper translation of dendritic mRNAs in response to synaptic activation. There are two different pathological FMR1 allele classes that are distinguished only by the number of repeats. Premutation alleles have 55-200 repeats and confer risk of fragile X-associated tremor/ataxia syndrome and fragile X-associated primary ovarian insufficiency. Full mutation alleles on the other hand have >200 repeats and result in fragile X syndrome, a disorder that affects learning and behavior. Different symptoms are seen in carriers of premutation and full mutation alleles because the repeat number has paradoxical effects on gene expression: Epigenetic changes increase transcription from premutation alleles and decrease transcription from full mutation alleles. This review will cover what is currently known about the mechanisms responsible for these changes in FMR1 expression and how they may relate to other Repeat Expansion Diseases that also show repeat-mediated changes in gene expression.
- Published
- 2015
- Full Text
- View/download PDF
94. Heterozygosity for a hypomorphic Polβ mutation reduces the expansion frequency in a mouse model of the Fragile X-related disorders.
- Author
-
Lokanga RA, Senejani AG, Sweasy JB, and Usdin K
- Subjects
- Animals, DNA Mismatch Repair genetics, Disease Models, Animal, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome pathology, Heterozygote, Humans, Mice, Mutation, DNA Polymerase beta genetics, DNA Repair genetics, Fragile X Syndrome genetics, Trinucleotide Repeat Expansion genetics
- Abstract
The Fragile X-related disorders (FXDs) are members of the Repeat Expansion Diseases, a group of human genetic conditions resulting from expansion of a specific tandem repeat. The FXDs result from expansion of a CGG/CCG repeat tract in the 5' UTR of the FMR1 gene. While expansion in a FXD mouse model is known to require some mismatch repair (MMR) proteins, our previous work and work in mouse models of another Repeat Expansion Disease show that early events in the base excision repair (BER) pathway play a role in the expansion process. One model for repeat expansion proposes that a non-canonical MMR process makes use of the nicks generated early in BER to load the MMR machinery that then generates expansions. However, we show here that heterozygosity for a Y265C mutation in Polβ, a key polymerase in the BER pathway, is enough to significantly reduce both the number of expansions seen in paternal gametes and the extent of somatic expansion in some tissues of the FXD mouse. These data suggest that events in the BER pathway downstream of the generation of nicks are also important for repeat expansion. Somewhat surprisingly, while the number of expansions is smaller, the average size of the residual expansions is larger than that seen in WT animals. This may have interesting implications for the mechanism by which BER generates expansions.
- Published
- 2015
- Full Text
- View/download PDF
95. The transcription-coupled repair protein ERCC6/CSB also protects against repeat expansion in a mouse model of the fragile X premutation.
- Author
-
Zhao XN and Usdin K
- Subjects
- Animals, DNA Repair Enzymes deficiency, Disease Models, Animal, Female, Gene Deletion, Genomic Instability, Genotype, Male, Mice, Mice, Knockout, MutS Homolog 2 Protein deficiency, Poly-ADP-Ribose Binding Proteins, DNA Repair Enzymes genetics, Fragile X Syndrome genetics, Transcription, Genetic, Trinucleotide Repeat Expansion
- Abstract
The fragile X-related disorders (FXDs) are members of the group of diseases known as the repeat expansion diseases. The FXDs result from expansion of an unstable CGG/CCG repeat tract in the 5' UTR of the FMR1 gene. Contractions are also seen, albeit at lower frequency. We have previously shown that ERCC6/CSB plays an auxiliary role in promoting germ line and somatic expansions in a mouse model of the FXDs. However, work in model systems of other repeat expansion diseases has suggested that CSB may protect against expansions by promoting contractions. Since FXD mice normally have such a high expansion frequency, it is possible that such a protective effect would have been masked. We thus examined the effect of the loss of CSB in an Msh2(+/-) background where the germ line expansion frequency is reduced and in an Msh2(-/-) background where expansions do not occur, but contractions do. Our data show that in addition to promoting repeat expansion, CSB does in fact protect the genome from germ line expansions in the FXD mouse model. However, it likely does so not by promoting contractions but by promoting an error-free process that preserves the parental allele., (Published 2015. Wiley Periodicals, Inc. **This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2015
- Full Text
- View/download PDF
96. Repeat instability during DNA repair: Insights from model systems.
- Author
-
Usdin K, House NC, and Freudenreich CH
- Subjects
- Chromosome Fragility genetics, DNA chemistry, DNA Damage, DNA Replication genetics, Genetic Diseases, Inborn classification, Genetic Diseases, Inborn etiology, Genomic Instability, Humans, Recombination, Genetic, DNA genetics, DNA Repair genetics, Nucleic Acid Conformation, Trinucleotide Repeat Expansion genetics
- Abstract
The expansion of repeated sequences is the cause of over 30 inherited genetic diseases, including Huntington disease, myotonic dystrophy (types 1 and 2), fragile X syndrome, many spinocerebellar ataxias, and some cases of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Repeat expansions are dynamic, and disease inheritance and progression are influenced by the size and the rate of expansion. Thus, an understanding of the various cellular mechanisms that cooperate to control or promote repeat expansions is of interest to human health. In addition, the study of repeat expansion and contraction mechanisms has provided insight into how repair pathways operate in the context of structure-forming DNA, as well as insights into non-canonical roles for repair proteins. Here we review the mechanisms of repeat instability, with a special emphasis on the knowledge gained from the various model systems that have been developed to study this topic. We cover the repair pathways and proteins that operate to maintain genome stability, or in some cases cause instability, and the cross-talk and interactions between them.
- Published
- 2015
- Full Text
- View/download PDF
97. Identification of fragile X syndrome specific molecular markers in human fibroblasts: a useful model to test the efficacy of therapeutic drugs.
- Author
-
Kumari D, Bhattacharya A, Nadel J, Moulton K, Zeak NM, Glicksman A, Dobkin C, Brick DJ, Schwartz PH, Smith CB, Klann E, and Usdin K
- Subjects
- Animals, Case-Control Studies, Cells, Cultured, Drug Evaluation, Preclinical, Fibroblasts cytology, Fibroblasts metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome drug therapy, Humans, Leucine metabolism, Male, Mice, Mice, Knockout, Phosphatidylinositol 3-Kinases metabolism, Protein Biosynthesis, RNA, Messenger genetics, Ribosomal Protein S6 Kinases metabolism, Biomarkers metabolism, Fragile X Syndrome genetics
- Abstract
Fragile X syndrome (FXS) is the most frequent cause of inherited intellectual disability and autism. It is caused by the absence of the fragile X mental retardation 1 (FMR1) gene product, fragile X mental retardation protein (FMRP), an RNA-binding protein involved in the regulation of translation of a subset of brain mRNAs. In Fmr1 knockout mice, the absence of FMRP results in elevated protein synthesis in the brain as well as increased signaling of many translational regulators. Whether protein synthesis is also dysregulated in FXS patients is not firmly established. Here, we demonstrate that fibroblasts from FXS patients have significantly elevated rates of basal protein synthesis along with increased levels of phosphorylated mechanistic target of rapamycin (p-mTOR), phosphorylated extracellular signal regulated kinase 1/2, and phosphorylated p70 ribosomal S6 kinase 1 (p-S6K1). The treatment with small molecules that inhibit S6K1 and a known FMRP target, phosphoinositide 3-kinase (PI3K) catalytic subunit p110β, lowered the rates of protein synthesis in both control and patient fibroblasts. Our data thus demonstrate that fibroblasts from FXS patients may be a useful in vitro model to test the efficacy and toxicity of potential therapeutics prior to clinical trials, as well as for drug screening and designing personalized treatment approaches., (© 2014 WILEY PERIODICALS, INC.)
- Published
- 2014
- Full Text
- View/download PDF
98. Repeat-mediated genetic and epigenetic changes at the FMR1 locus in the Fragile X-related disorders.
- Author
-
Usdin K, Hayward BE, Kumari D, Lokanga RA, Sciascia N, and Zhao XN
- Abstract
The Fragile X-related disorders are a group of genetic conditions that include the neurodegenerative disorder, Fragile X-associated tremor/ataxia syndrome (FXTAS), the fertility disorder, Fragile X-associated primary ovarian insufficiency (FXPOI) and the intellectual disability, Fragile X syndrome (FXS). The pathology in all these diseases is related to the number of CGG/CCG-repeats in the 5' UTR of the Fragile X mental retardation 1 (FMR1) gene. The repeats are prone to continuous expansion and the increase in repeat number has paradoxical effects on gene expression increasing transcription on mid-sized alleles and decreasing it on longer ones. In some cases the repeats can simultaneously both increase FMR1 mRNA production and decrease the levels of the FMR1 gene product, Fragile X mental retardation 1 protein (FMRP). Since FXTAS and FXPOI result from the deleterious consequences of the expression of elevated levels of FMR1 mRNA and FXS is caused by an FMRP deficiency, the clinical picture is turning out to be more complex than once appreciated. Added complications result from the fact that increasing repeat numbers make the alleles somatically unstable. Thus many individuals have a complex mixture of different sized alleles in different cells. Furthermore, it has become apparent that the eponymous fragile site, once thought to be no more than a useful diagnostic criterion, may have clinical consequences for females who inherit chromosomes that express this site. This review will cover what is currently known about the mechanisms responsible for repeat instability, for the repeat-mediated epigenetic changes that affect expression of the FMR1 gene, and for chromosome fragility. It will also touch on what current and future options are for ameliorating some of these effects.
- Published
- 2014
- Full Text
- View/download PDF
99. Chromosome fragility and the abnormal replication of the FMR1 locus in fragile X syndrome.
- Author
-
Yudkin D, Hayward BE, Aladjem MI, Kumari D, and Usdin K
- Subjects
- Cell Line, Chromosome Fragile Sites, Chromosomes, Human, X chemistry, Chromosomes, Human, X genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome metabolism, Heterozygote, Humans, Trinucleotide Repeats, Chromosome Fragility, DNA Replication, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics
- Abstract
Fragile X Syndrome (FXS) is a learning disability seen in individuals who have >200 CGG•CCG repeats in the 5' untranslated region of the X-linked FMR1 gene. Such alleles are associated with a fragile site, FRAXA, a gap or constriction in the chromosome that is coincident with the repeat and is induced by folate stress or thymidylate synthase inhibitors like fluorodeoxyuridine (FdU). The molecular basis of the chromosome fragility is unknown. Previous work has suggested that the stable intrastrand structures formed by the repeat may be responsible, perhaps via their ability to block DNA synthesis. We have examined the replication dynamics of normal and FXS cells with and without FdU. We show here that an intrinsic problem with DNA replication exists in the FMR1 gene of individuals with FXS even in the absence of FdU. Our data suggest a model for chromosome fragility in FXS in which the repeat impairs replication from an origin of replication (ORI) immediately adjacent to the repeat. The fact that the replication problem occurs even in the absence of FdU suggests that this phenomenon may have in vivo consequences, including perhaps accounting for the loss of the X chromosome containing the fragile site that causes Turner syndrome (45, X0) in female carriers of such alleles. Our data on FRAXA may also be germane for the other FdU-inducible fragile sites in humans, that we show here share many common features with FRAXA.
- Published
- 2014
- Full Text
- View/download PDF
100. Gender and cell-type-specific effects of the transcription-coupled repair protein, ERCC6/CSB, on repeat expansion in a mouse model of the fragile X-related disorders.
- Author
-
Zhao XN and Usdin K
- Subjects
- 5' Untranslated Regions, Alleles, Animals, Disease Models, Animal, Female, Fragile X Mental Retardation Protein genetics, Male, Mice, Mice, Knockout, Poly-ADP-Ribose Binding Proteins, RNA, Messenger genetics, DNA Repair, DNA Repair Enzymes genetics, DNA Repeat Expansion, Fragile X Syndrome genetics, Sex Factors, Transcription, Genetic
- Abstract
The repeat expansion diseases are human genetic disorders that arise from the expansion of a tandem-repeat tract. The Fragile X-related disorders are members of this disease group in which the repeat unit is CGG/CCG and is located in the 5′ untranslated region of the FMR1 gene. Affected individuals often show mosaicism with respect to repeat number resulting from both expansion and contraction of the repeat tract; however, the mechanism responsible for these changes in repeat number is unknown. The work from a variety of model systems suggests that transcription-coupled repair (TCR) may contribute to repeat instability in diseases resulting from CAG/CTG-repeat expansion. To test whether TCR could contribute to repeat instability in the Fragile X-related disorders, we tested the effect of mutations in Csb (Cockayne syndrome group B), a gene essential for TCR, in a knock-in mouse model of these disorders. We found that the loss of CSB affects expansions in a gender and cell-type-specific manner. Our data also show an unanticipated gender difference in instability even in Csb+/+ animals that may have implications for our understanding of the mechanism of repeat expansion in the FX mouse model and perhaps for humans as well.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.