89 results on '"Todd PK"'
Search Results
2. Stress granule formation helps to mitigate neurodegeneration.
- Author
-
Glineburg MR, Yildirim E, Gomez N, Rodriguez G, Pak J, Li X, Altheim C, Waksmacki J, McInerney GM, Barmada SJ, and Todd PK
- Subjects
- Animals, Humans, RNA Recognition Motif Proteins metabolism, RNA Recognition Motif Proteins genetics, Drosophila Proteins metabolism, Drosophila Proteins genetics, Poly-ADP-Ribose Binding Proteins metabolism, Poly-ADP-Ribose Binding Proteins genetics, Mice, Drosophila melanogaster metabolism, Drosophila melanogaster genetics, RNA Helicases metabolism, RNA Helicases genetics, Ataxia genetics, Ataxia metabolism, DNA Helicases metabolism, DNA Helicases genetics, Alphavirus genetics, Alphavirus metabolism, Rats, Carrier Proteins metabolism, Drosophila metabolism, Cytoplasmic Granules metabolism, Stress, Physiological, DNA-Binding Proteins, Stress Granules metabolism, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases genetics, Amyotrophic Lateral Sclerosis metabolism, Amyotrophic Lateral Sclerosis genetics, Neurons metabolism, Frontotemporal Dementia metabolism, Frontotemporal Dementia genetics
- Abstract
Cellular stress pathways that inhibit translation initiation lead to transient formation of cytoplasmic RNA/protein complexes known as stress granules. Many of the proteins found within stress granules and the dynamics of stress granule formation and dissolution are implicated in neurodegenerative disease. Whether stress granule formation is protective or harmful in neurodegenerative conditions is not known. To address this, we took advantage of the alphavirus protein nsP3, which selectively binds dimers of the central stress granule nucleator protein G3BP and markedly reduces stress granule formation without directly impacting the protein translational inhibitory pathways that trigger stress granule formation. In Drosophila and rodent neurons, reducing stress granule formation with nsP3 had modest impacts on lifespan even in the setting of serial stress pathway induction. In contrast, reducing stress granule formation in models of ataxia, amyotrophic lateral sclerosis and frontotemporal dementia largely exacerbated disease phenotypes. These data support a model whereby stress granules mitigate, rather than promote, neurodegenerative cascades., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
3. AAGGG repeat expansions trigger RFC1 -independent synaptic dysregulation in human CANVAS neurons.
- Author
-
Maltby CJ, Krans A, Grudzien SJ, Palacios Y, Muiños J, Suárez A, Asher M, Willey S, Van Deynze K, Mumm C, Boyle AP, Cortese A, Ndayisaba A, Khurana V, Barmada SJ, Dijkstra AA, and Todd PK
- Subjects
- Humans, Bilateral Vestibulopathy genetics, Bilateral Vestibulopathy metabolism, Vestibular Diseases genetics, Alleles, Replication Protein C genetics, Replication Protein C metabolism, Neurons metabolism, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells cytology, DNA Repeat Expansion genetics, Cerebellar Ataxia genetics, Cerebellar Ataxia pathology, Cerebellar Ataxia metabolism, Synapses metabolism, Synapses genetics
- Abstract
Cerebellar ataxia with neuropathy and vestibular areflexia syndrome (CANVAS) is a recessively inherited neurodegenerative disorder caused by intronic biallelic, nonreference CCCTT/AAGGG repeat expansions within RFC1 . To investigate how these repeats cause disease, we generated patient induced pluripotent stem cell-derived neurons (iNeurons). CCCTT/AAGGG repeat expansions do not alter neuronal RFC1 splicing, expression, or DNA repair pathway function. In reporter assays, AAGGG repeats are translated into pentapeptide repeat proteins. However, these proteins and repeat RNA foci were not detected in iNeurons, and overexpression of these repeats failed to induce neuronal toxicity. CANVAS iNeurons exhibit defects in neuronal development and diminished synaptic connectivity that is rescued by CRISPR deletion of a single expanded AAGGG allele. These deficits were neither replicated by RFC1 knockdown in control iNeurons nor rescued by RFC1 reprovision in CANVAS iNeurons. These findings support a repeat-dependent but RFC1 protein-independent cause of neuronal dysfunction in CANVAS, with implications for therapeutic development in this currently untreatable condition.
- Published
- 2024
- Full Text
- View/download PDF
4. Ribosomal quality control factors inhibit repeat-associated non-AUG translation from GC-rich repeats.
- Author
-
Tseng YJ, Krans A, Malik I, Deng X, Yildirim E, Ovunc S, Tank EMH, Jansen-West K, Kaufhold R, Gomez NB, Sher R, Petrucelli L, Barmada SJ, and Todd PK
- Subjects
- Humans, Ataxia, DNA Repeat Expansion genetics, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome genetics, Fragile X Syndrome metabolism, GC Rich Sequence, HEK293 Cells, Induced Pluripotent Stem Cells metabolism, Neurons metabolism, Ribosomes metabolism, Ribosomes genetics, Tremor, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, C9orf72 Protein genetics, C9orf72 Protein metabolism, Frontotemporal Dementia genetics, Frontotemporal Dementia metabolism, Protein Biosynthesis, Trinucleotide Repeat Expansion genetics, Ribosomal Proteins metabolism
- Abstract
A GGGGCC (G4C2) hexanucleotide repeat expansion in C9ORF72 causes amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD), while a CGG trinucleotide repeat expansion in FMR1 leads to the neurodegenerative disorder Fragile X-associated tremor/ataxia syndrome (FXTAS). These GC-rich repeats form RNA secondary structures that support repeat-associated non-AUG (RAN) translation of toxic proteins that contribute to disease pathogenesis. Here we assessed whether these same repeats might trigger stalling and interfere with translational elongation. We find that depletion of ribosome-associated quality control (RQC) factors NEMF, LTN1 and ANKZF1 markedly boost RAN translation product accumulation from both G4C2 and CGG repeats while overexpression of these factors reduces RAN production in both reporter assays and C9ALS/FTD patient iPSC-derived neurons. We also detected partially made products from both G4C2 and CGG repeats whose abundance increased with RQC factor depletion. Repeat RNA sequence, rather than amino acid content, is central to the impact of RQC factor depletion on RAN translation-suggesting a role for RNA secondary structure in these processes. Together, these findings suggest that ribosomal stalling and RQC pathway activation during RAN translation inhibits the generation of toxic RAN products. We propose augmenting RQC activity as a therapeutic strategy in GC-rich repeat expansion disorders., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
5. Enhanced Detection and Genotyping of Disease-Associated Tandem Repeats Using HMMSTR and Targeted Long-Read Sequencing.
- Author
-
Van Deynze K, Mumm C, Maltby CJ, Switzenberg JA, Todd PK, and Boyle AP
- Abstract
Tandem repeat sequences comprise approximately 8% of the human genome and are linked to more than 50 neurodegenerative disorders. Accurate characterization of disease-associated repeat loci remains resource intensive and often lacks high resolution genotype calls. We introduce a multiplexed, targeted nanopore sequencing panel and HMMSTR, a sequence-based tandem repeat copy number caller. HMMSTR outperforms current signal- and sequence-based callers relative to two assemblies and we show it performs with high accuracy in heterozygous regions and at low read coverage. The flexible panel allows us to capture disease associated regions at an average coverage of >150x. Using these tools, we successfully characterize known or suspected repeat expansions in patient derived samples. In these samples we also identify unexpected expanded alleles at tandem repeat loci not previously associated with the underlying diagnosis. This genotyping approach for tandem repeat expansions is scalable, simple, flexible, and accurate, offering significant potential for diagnostic applications and investigation of expansion co-occurrence in neurodegenerative disorders.
- Published
- 2024
- Full Text
- View/download PDF
6. AAGGG repeat expansions trigger RFC1-independent synaptic dysregulation in human CANVAS Neurons.
- Author
-
Maltby CJ, Krans A, Grudzien SJ, Palacios Y, Muiños J, Suárez A, Asher M, Khurana V, Barmada SJ, Dijkstra AA, and Todd PK
- Abstract
Cerebellar ataxia with neuropathy and vestibular areflexia syndrome (CANVAS) is a late onset, recessively inherited neurodegenerative disorder caused by biallelic, non-reference pentameric AAGGG(CCCTT) repeat expansions within the second intron of replication factor complex subunit 1 ( RFC1 ). To investigate how these repeats cause disease, we generated CANVAS patient induced pluripotent stem cell (iPSC) derived neurons (iNeurons) and utilized calcium imaging and transcriptomic analysis to define repeat-elicited gain-of-function and loss-of-function contributions to neuronal toxicity. AAGGG repeat expansions do not alter neuronal RFC1 splicing, expression, or DNA repair pathway functions. In reporter assays, AAGGG repeats are translated into pentapeptide repeat proteins that selectively accumulate in CANVAS patient brains. However, neither these proteins nor repeat RNA foci were detected in iNeurons, and overexpression of these repeats in isolation did not induce neuronal toxicity. CANVAS iNeurons exhibit defects in neuronal development and diminished synaptic connectivity that is rescued by CRISPR deletion of a single expanded allele. These phenotypic deficits were not replicated by knockdown of RFC1 in control neurons and were not rescued by ectopic expression of RFC1. These findings support a repeat-dependent but RFC1-independent cause of neuronal dysfunction in CANVAS, with important implications for therapeutic development in this currently untreatable condition., Competing Interests: Conflicts of interest The authors declare no direct conflicts of interest related to the content of this manuscript. No commercial forces had editorial or supervisory input on the content of the manuscript or its figures. P.K.T. holds a shared patent on ASOs with Ionis Pharmaceuticals. He has served as a consultant with Denali Therapeutics, and he has licensed technology and antibodies to Denali and Abcam. V.K. is a co-founder of and senior advisor to DaCapo Brainscience and Yumanity Therapeutics, companies focused on CNS diseases.
- Published
- 2023
- Full Text
- View/download PDF
7. Stress granule formation helps to mitigate neurodegeneration.
- Author
-
Glineburg MR, Yildirim E, Gomez N, Li X, Pak J, Altheim C, Waksmacki J, McInerney G, Barmada SJ, and Todd PK
- Abstract
Cellular stress pathways that inhibit translation initiation lead to transient formation of cytoplasmic RNA/protein complexes known as stress granules. Many of the proteins found within stress granules and the dynamics of stress granule formation and dissolution are implicated in neurodegenerative disease. Whether stress granule formation is protective or harmful in neurodegenerative conditions is not known. To address this, we took advantage of the alphavirus protein nsP3, which selectively binds dimers of the central stress granule nucleator protein G3BP ( rin in Drosophila ) and markedly reduces stress granule formation without directly impacting the protein translational inhibitory pathways that trigger stress granule formation. In Drosophila and rodent neurons, reducing stress granule formation with nsP3 had modest impacts on lifespan even in the setting of serial stress pathway induction. In contrast, reducing stress granule formation in models of ataxia, amyotrophic lateral sclerosis and frontotemporal dementia largely exacerbated disease phenotypes. These data support a model whereby stress granules mitigate, rather than promote, neurodegenerative cascades.
- Published
- 2023
- Full Text
- View/download PDF
8. Insight and Recommendations for Fragile X-Premutation-Associated Conditions from the Fifth International Conference on FMR1 Premutation.
- Author
-
Tassone F, Protic D, Allen EG, Archibald AD, Baud A, Brown TW, Budimirovic DB, Cohen J, Dufour B, Eiges R, Elvassore N, Gabis LV, Grudzien SJ, Hall DA, Hessl D, Hogan A, Hunter JE, Jin P, Jiraanont P, Klusek J, Kooy RF, Kraan CM, Laterza C, Lee A, Lipworth K, Losh M, Loesch D, Lozano R, Mailick MR, Manolopoulos A, Martinez-Cerdeno V, McLennan Y, Miller RM, Montanaro FAM, Mosconi MW, Potter SN, Raspa M, Rivera SM, Shelly K, Todd PK, Tutak K, Wang JY, Wheeler A, Winarni TI, Zafarullah M, and Hagerman RJ
- Subjects
- Humans, Mutation genetics, RNA, Messenger metabolism, Trinucleotide Repeat Expansion genetics, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome diagnosis, Fragile X Syndrome genetics, Fragile X Syndrome therapy
- Abstract
The premutation of the fragile X messenger ribonucleoprotein 1 ( FMR1 ) gene is characterized by an expansion of the CGG trinucleotide repeats (55 to 200 CGGs) in the 5' untranslated region and increased levels of FMR1 mRNA. Molecular mechanisms leading to fragile X-premutation-associated conditions (FXPAC) include cotranscriptional R-loop formations, FMR1 mRNA toxicity through both RNA gelation into nuclear foci and sequestration of various CGG-repeat-binding proteins, and the repeat-associated non-AUG (RAN)-initiated translation of potentially toxic proteins. Such molecular mechanisms contribute to subsequent consequences, including mitochondrial dysfunction and neuronal death. Clinically, premutation carriers may exhibit a wide range of symptoms and phenotypes. Any of the problems associated with the premutation can appropriately be called FXPAC. Fragile X-associated tremor/ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), and fragile X-associated neuropsychiatric disorders (FXAND) can fall under FXPAC. Understanding the molecular and clinical aspects of the premutation of the FMR1 gene is crucial for the accurate diagnosis, genetic counseling, and appropriate management of affected individuals and families. This paper summarizes all the known problems associated with the premutation and documents the presentations and discussions that occurred at the International Premutation Conference, which took place in New Zealand in 2023.
- Published
- 2023
- Full Text
- View/download PDF
9. Dissecting the roles of EIF4G homologs reveals DAP5 as a modifier of CGG repeat-associated toxicity in a Drosophila model of FXTAS.
- Author
-
Malik I, Tseng YJ, Wieland CM, Green KM, Zheng K, Calleja K, and Todd PK
- Subjects
- Animals, Drosophila metabolism, Tremor genetics, Drosophila melanogaster metabolism, Eukaryotic Initiation Factor-4G genetics, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Trinucleotide Repeat Expansion, Ataxia genetics, Mammals metabolism, Fragile X Syndrome genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism
- Abstract
Neurodegeneration in Fragile X-associated tremor/ataxia syndrome (FXTAS) is caused by a CGG trinucleotide repeat expansion in the 5' UTR of FMR1. Expanded CGG repeat RNAs form stable secondary structures, which in turn support repeat-associated non-AUG (RAN) translation to produce toxic peptides. The parameters that impact RAN translation initiation efficiency are not well understood. Here we used a Drosophila melanogaster model of FXTAS to evaluate the role of the eIF4G family of eukaryotic translation initiation factors (EIF4G1, EIF4GII and EIF4G2/DAP5) in modulating RAN translation and CGG repeat-associated toxicity. DAP5 knockdown robustly suppressed CGG repeat-associated toxicity and inhibited RAN translation. Furthermore, knockdown of initiation factors that preferentially associate with DAP5 (such as EIF2β, EIF3F and EIF3G) also selectively suppressed CGG repeat-induced eye degeneration. In mammalian cellular reporter assays, DAP5 knockdown exhibited modest and cell-type specific effects on RAN translation. Taken together, these data support a role for DAP5 in CGG repeat associated toxicity possibly through modulation of RAN translation., Competing Interests: Declaration of Competing Interest The authors have no financial conflicts of interest associated with the work presented in this manuscript., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
10. Ribosomal quality control factors inhibit repeat-associated non-AUG translation from GC-rich repeats.
- Author
-
Tseng YJ, Malik I, Deng X, Krans A, Jansen-West K, Tank EMH, Gomez NB, Sher R, Petrucelli L, Barmada SJ, and Todd PK
- Abstract
A GGGGCC (G4C2) hexanucleotide repeat expansion in C9ORF72 causes amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD), while a CGG trinucleotide repeat expansion in FMR1 leads to the neurodegenerative disorder Fragile X-associated tremor/ataxia syndrome (FXTAS). These GC-rich repeats form RNA secondary structures that support repeat-associated non-AUG (RAN) translation of toxic proteins that contribute to disease pathogenesis. Here we assessed whether these same repeats might trigger stalling and interfere with translational elongation. We find that depletion of ribosome-associated quality control (RQC) factors NEMF, LTN1, and ANKZF1 markedly boost RAN translation product accumulation from both G4C2 and CGG repeats while overexpression of these factors reduces RAN production in both reporter cell lines and C9ALS/FTD patient iPSC-derived neurons. We also detected partially made products from both G4C2 and CGG repeats whose abundance increased with RQC factor depletion. Repeat RNA sequence, rather than amino acid content, is central to the impact of RQC factor depletion on RAN translation - suggesting a role for RNA secondary structure in these processes. Together, these findings suggest that ribosomal stalling and RQC pathway activation during RAN translation elongation inhibits the generation of toxic RAN products. We propose augmenting RQC activity as a therapeutic strategy in GC-rich repeat expansion disorders., Competing Interests: CONFLICT OF INTEREST The authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
11. Native functions of short tandem repeats.
- Author
-
Wright SE and Todd PK
- Subjects
- Humans, Gene Expression Regulation, Microsatellite Repeats genetics, Genome, Human
- Abstract
Over a third of the human genome is comprised of repetitive sequences, including more than a million short tandem repeats (STRs). While studies of the pathologic consequences of repeat expansions that cause syndromic human diseases are extensive, the potential native functions of STRs are often ignored. Here, we summarize a growing body of research into the normal biological functions for repetitive elements across the genome, with a particular focus on the roles of STRs in regulating gene expression. We propose reconceptualizing the pathogenic consequences of repeat expansions as aberrancies in normal gene regulation. From this altered viewpoint, we predict that future work will reveal broader roles for STRs in neuronal function and as risk alleles for more common human neurological diseases., Competing Interests: SW No competing interests declared, PT Dr Todd served as a consultant to Denali Therapeutics and holds a shared patent on ASOs developed with Ionis Pharmaceuticals
- Published
- 2023
- Full Text
- View/download PDF
12. What repeat expansion disorders can teach us about the Central Dogma.
- Author
-
Wang ET, Freudenreich CH, Gromak N, Jain A, Todd PK, and Nagai Y
- Subjects
- Humans, Proteins genetics, RNA genetics, Trinucleotide Repeat Expansion genetics, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis pathology, Huntington Disease genetics, Myotonic Dystrophy genetics
- Abstract
Pathogenic repeat sequences underlie several human disorders, including amyotrophic lateral sclerosis, Huntington's disease, and myotonic dystrophy. Here, we speak to several researchers about how repeat sequences have been implicated in affecting all aspects of the Central Dogma of molecular biology through their effects on DNA, RNA, and protein., Competing Interests: Declaration of interests E.T.W. is a cofounder of and consultant to Kate Therapeutics. A.J. is a member of the scientific advisory board of Molecular Cell. P.K.T. served as a consultant to Denali Therapeutics and holds a shared patent with Ionis Pharmaceuticals. Y.N. discloses the patent PCT/JP2017/23,162 related to this work., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. The International Fragile X Premutation Registry: building a resource for research and clinical trial readiness.
- Author
-
Hessl D, Rosselot H, Miller R, Espinal G, Famula J, Sherman SL, Todd PK, Cabal Herrera AM, Lipworth K, Cohen J, Hall DA, Leehey M, Grigsby J, Weber JD, Alusi S, Wheeler A, Raspa M, Hudson T, and Sobrian SK
- Subjects
- Humans, Trinucleotide Repeat Expansion genetics, Registries, Guanine, Fragile X Mental Retardation Protein genetics, Neurodegenerative Diseases genetics
- Abstract
FMR1 premutation cytosine-guanine-guanine repeat expansion alleles are relatively common mutations in the general population that are associated with a neurodegenerative disease (fragile X-associated tremor/ataxia syndrome), reproductive health problems and potentially a wide range of additional mental and general health conditions that are not yet well-characterised. The International Fragile X Premutation Registry (IFXPR) was developed to facilitate and encourage research to better understand the FMR1 premutation and its impact on human health, to facilitate clinical trial readiness by identifying and characterising diverse cohorts of individuals interested in study participation, and to build community and collaboration among carriers, family members, researchers and clinicians around the world. Here, we describe the development and content of the IFXPR, characterise its first 747 registrants from 32 countries and invite investigators to apply for recruitment support for their project(s). With larger numbers, increased diversity and potentially the future clinical characterisation of registrants, the IFXPR will contribute to a more comprehensive and accurate understanding of the fragile X premutation in human health and support treatment studies., Competing Interests: Competing interests: DH is a member of the Clinical and Scientific Advisory Committee and the Clinical Trials Committee of the National Fragile X Foundation., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
14. Repeat-Associated Non-AUG Translation of AGAGGG Repeats that Cause X-Linked Dystonia-Parkinsonism.
- Author
-
Reyes CJ, Asano K, Todd PK, Klein C, and Rakovic A
- Subjects
- Humans, HEK293 Cells, Introns, C9orf72 Protein genetics, Dystonic Disorders metabolism, Genetic Diseases, X-Linked genetics
- Abstract
Background: X-linked dystonia-parkinsonism (XDP) is a neurodegenerative disorder caused by the intronic insertion of a SINE-VNTR-Alu (SVA) retrotransposon carrying an (AGAGGG)
n repeat expansion in the TAF1 gene. The molecular mechanisms by which this mutation causes neurodegeneration remain elusive., Objectives: We investigated whether (AGAGGG)n repeats undergo repeat-associated non-AUG (RAN) translation, a pathogenic mechanism common among repeat expansion diseases., Methods: XDP-specific RAN translation reporter plasmids were generated, transfected in HEK293 cells, and putative dipeptide repeat proteins (DPRs) were detected by Western blotting. Immunocytochemistry was performed in COS-7 cells to determine the subcellular localization of one DPR., Results: We detected putative DPRs from two reading frames, supporting the translation of poly-(Glu-Gly) and poly-(Arg-Glu) species. XDP RAN translation initiates within the (AGAGGG)n sequence and poly-(Glu-Gly) DPRs formed nuclear inclusions in transfected cells., Conclusions: In summary, our work provides the first in-vitro proof of principle that the XDP-linked (AGAGGG)n repeat expansions can undergo RAN translation. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society., (© 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.)- Published
- 2022
- Full Text
- View/download PDF
15. Fragile X-associated tremor ataxia syndrome rating scale: Revision and content validity using a mixed method approach.
- Author
-
Tosin MHS, Stebbins GT, Goetz CG, Hagerman RJ, Hessl D, Zolecki MA, Todd PK, Leehey MA, and Hall DA
- Abstract
Background: The original Fragile X-associated Tremor Ataxia Syndrome Rating Scale (FXTAS-RS) contained 61 items, some requiring modifications to better meet recommendations for patient-focused rating scale development., Purpose: Provide initial validation of a revised version of the FXTAS-RS for motor signs., Method: We conducted a two-phase mixed-method approach. In Phase 1, revision, we implemented a Delphi technique identifying pertinent domains/subdomains and developing items through expert consensus. In Phase 2, content validation, we conducted cognitive pretesting assessing comprehensibility, comprehensiveness, and relevance of items to FXTAS motor signs., Results: After five rounds of Delphi panel and two rounds of cognitive pretesting, the revised version of the FXTAS-RS was established with 18 items covering five domains and 13 subdomains of motor signs. Cognitive pretesting revealed adequate content validity for the assessment of FXTAS motor signs., Conclusion: The revised FXTAS-RS has been successfully validated for content and it is now ready for large-scale field validation., 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 © 2022 Tosin, Stebbins, Goetz, Hagerman, Hessl, Zolecki, Todd, Leehey and Hall.)
- Published
- 2022
- Full Text
- View/download PDF
16. CGG repeats trigger translational frameshifts that generate aggregation-prone chimeric proteins.
- Author
-
Wright SE, Rodriguez CM, Monroe J, Xing J, Krans A, Flores BN, Barsur V, Ivanova MI, Koutmou KS, Barmada SJ, and Todd PK
- Subjects
- Arginine genetics, Ataxia, Fragile X Syndrome, Glycine genetics, Humans, Peptides genetics, Peptides metabolism, Recombinant Fusion Proteins metabolism, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Neurodegenerative Diseases genetics, Protein Aggregation, Pathological, Trinucleotide Repeats
- Abstract
CGG repeat expansions in the FMR1 5'UTR cause the neurodegenerative disease Fragile X-associated tremor/ataxia syndrome (FXTAS). These repeats form stable RNA secondary structures that support aberrant translation in the absence of an AUG start codon (RAN translation), producing aggregate-prone peptides that accumulate within intranuclear neuronal inclusions and contribute to neurotoxicity. Here, we show that the most abundant RAN translation product, FMRpolyG, is markedly less toxic when generated from a construct with a non-repetitive alternating codon sequence in place of the CGG repeat. While exploring the mechanism of this differential toxicity, we observed a +1 translational frameshift within the CGG repeat from the arginine to glycine reading frame. Frameshifts occurred within the first few translated repeats and were triggered predominantly by RNA sequence and structural features. Short chimeric R/G peptides form aggregates distinct from those formed by either pure arginine or glycine, and these chimeras induce toxicity in cultured rodent neurons. Together, this work suggests that CGG repeats support translational frameshifting and that chimeric RAN translated peptides may contribute to CGG repeat-associated toxicity in FXTAS and related disorders., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
17. Non-canonical initiation factors modulate repeat-associated non-AUG translation.
- Author
-
Green KM, Miller SL, Malik I, and Todd PK
- Subjects
- Animals, Drosophila genetics, Drosophila metabolism, Eukaryotic Initiation Factor-2 genetics, Eukaryotic Initiation Factor-2 metabolism, Eukaryotic Initiation Factors genetics, Eukaryotic Initiation Factors metabolism, Protein Biosynthesis genetics, RNA, Messenger genetics, Ribosomes genetics, Ribosomes metabolism, Drosophila Proteins genetics, Drosophila Proteins metabolism, Neurodegenerative Diseases genetics, Neurodegenerative Diseases metabolism
- Abstract
Repeat-associated non-AUG (RAN) translation of expanded repeat-mutation mRNA produces toxic peptides in neurons of patients suffering from neurodegenerative diseases. Recent findings indicate that RAN translation in diverse model systems is not inhibited by cellular stressors that impair global translation through phosphorylation of the alpha subunit of eIF2, the essential eukaryotic translation initiation factor that brings the initiator tRNA to the 40S ribosome. Using in vitro, cell-based and Drosophila models, we examined the role of alternative ternary complex factors that may function in place of eIF2, including eIF2A, eIF2D, DENR and MCTS1. Among these factors, DENR knockdown had the greatest inhibitory effect on RAN translation of expanded GGGGCC and CGG repeat reporters and its reduction improved the survival of Drosophila expressing expanded GGGGCC repeats. Taken together, these data support a role for alternative initiation factors in RAN translation and suggest these may serve as novel therapeutic targets in neurodegenerative disease., (Published by Oxford University Press 2022.)
- Published
- 2022
- Full Text
- View/download PDF
18. Mechanistic convergence across initiation sites for RAN translation in fragile X associated tremor ataxia syndrome.
- Author
-
Zhang Y, Glineburg MR, Basrur V, Conlon K, Wright SE, Krans A, Hall DA, and Todd PK
- Subjects
- Fragile X Mental Retardation Protein genetics, Humans, Peptides metabolism, Trinucleotide Repeat Expansion, Ataxia genetics, Fragile X Syndrome genetics, Tremor genetics, ran GTP-Binding Protein genetics
- Abstract
Repeat associated non-AUG (RAN) translation of CGG repeats in the 5'UTR of FMR1 produces toxic proteins that contribute to fragile X-associated tremor/ataxia syndrome (FXTAS) pathogenesis. The most abundant RAN product, FMRpolyG, initiates predominantly at an ACG upstream of the repeat. Accurate FMRpolyG measurements in FXTAS patients are lacking. We used data-dependent acquisition and parallel reaction monitoring (PRM) mass spectrometry coupled with stable isotope labeled standard peptides to identify signature FMRpolyG fragments in patient samples. Following immunoprecipitation, PRM detected FMRpolyG signature peptides in transfected cells, and FXTAS tissues and cells, but not in controls. We identified two amino-terminal peptides: an ACG-initiated Ac-MEAPLPGGVR and a GUG-initiated Ac-TEAPLPGGVR, as well as evidence for RAN translation initiation within the CGG repeat itself in two reading frames. Initiation at all sites increased following cellular stress, decreased following eIF1 overexpression and was eIF4A and M7G cap-dependent. These data demonstrate that FMRpolyG is quantifiable in human samples and FMR1 RAN translation initiates via similar mechanisms for near-cognate codons and within the repeat through processes dependent on available initiation factors and cellular environment., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
19. Identification of PSMB5 as a genetic modifier of fragile X-associated tremor/ataxia syndrome.
- Author
-
Kong HE, Lim J, Linsalata A, Kang Y, Malik I, Allen EG, Cao Y, Shubeck L, Johnston R, Huang Y, Gu Y, Guo X, Zwick ME, Qin Z, Wingo TS, Juncos J, Nelson DL, Epstein MP, Cutler DJ, Todd PK, Sherman SL, Warren ST, and Jin P
- Subjects
- Animals, Disease Models, Animal, Drosophila melanogaster, Fragile X Mental Retardation Protein genetics, Humans, Male, Ataxia genetics, Fragile X Syndrome genetics, Proteasome Endopeptidase Complex genetics, Tremor genetics
- Abstract
Fragile X–associated tremor/ataxia syndrome (FXTAS) is a debilitating late-onset neurodegenerative disease in premutation carriers of the expanded CGG repeat in FMR1 that presents with a spectrum of neurological manifestations, such as gait ataxia, intention tremor, and parkinsonism [P. J. Hagerman, R. J. Hagerman, Ann. N. Y. Acad. Sci. 1338, 58–70 (2015); S. Jacquemont et al., JAMA 291, 460–469 (2004)]. Here, we performed whole-genome sequencing (WGS) on male premutation carriers (CGG55–200) and prioritized candidate variants to screen for candidate genetic modifiers using a Drosophila model of FXTAS. We found 18 genes that genetically modulate CGG-associated neurotoxicity in Drosophila, such as Prosbeta5 (PSMB5), pAbp (PABPC1L), e(y)1 (TAF9), and CG14231 (OSGEPL1). Among them, knockdown of Prosbeta5 (PSMB5) suppressed CGG-associated neurodegeneration in the fly as well as in N2A cells. Interestingly, an expression quantitative trait locus variant in PSMB5, PSMB5rs11543947-A, was found to be associated with decreased expression of PSMB5 and delayed onset of FXTAS in human FMR1 premutation carriers. Finally, we demonstrate evidence that PSMB5 knockdown results in suppression of CGG neurotoxicity via both the RAN translation and RNA-mediated toxicity mechanisms, thereby presenting a therapeutic strategy for FXTAS.
- Published
- 2022
- Full Text
- View/download PDF
20. Reuterin in the healthy gut microbiome suppresses colorectal cancer growth through altering redox balance.
- Author
-
Bell HN, Rebernick RJ, Goyert J, Singhal R, Kuljanin M, Kerk SA, Huang W, Das NK, Andren A, Solanki S, Miller SL, Todd PK, Fearon ER, Lyssiotis CA, Gygi SP, Mancias JD, and Shah YM
- Subjects
- Animals, Biomarkers, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Energy Metabolism, Glutathione metabolism, Glyceraldehyde metabolism, Glyceraldehyde pharmacology, Host Microbial Interactions, Humans, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Intestinal Mucosa pathology, Metabolomics methods, Metagenomics methods, Mice, Models, Biological, Oxidative Stress, Propane pharmacology, Signal Transduction, Xenograft Model Antitumor Assays, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Gastrointestinal Microbiome, Glyceraldehyde analogs & derivatives, Oxidation-Reduction drug effects, Propane metabolism
- Abstract
Microbial dysbiosis is a colorectal cancer (CRC) hallmark and contributes to inflammation, tumor growth, and therapy response. Gut microbes signal via metabolites, but how the metabolites impact CRC is largely unknown. We interrogated fecal metabolites associated with mouse models of colon tumorigenesis with varying mutational load. We find that microbial metabolites from healthy mice or humans are growth-repressive, and this response is attenuated in mice and patients with CRC. Microbial profiling reveals that Lactobacillus reuteri and its metabolite, reuterin, are downregulated in mouse and human CRC. Reuterin alters redox balance, and reduces proliferation and survival in colon cancer cells. Reuterin induces selective protein oxidation and inhibits ribosomal biogenesis and protein translation. Exogenous Lactobacillus reuteri restricts colon tumor growth, increases tumor reactive oxygen species, and decreases protein translation in vivo. Our findings indicate that a healthy microbiome and specifically, Lactobacillus reuteri, is protective against CRC through microbial metabolite exchange., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. Guillain-Barré Syndrome After COVID-19 mRNA Vaccination in a Liver Transplantation Recipient With Favorable Treatment Response.
- Author
-
Hughes DL, Brunn JA, Jacobs J, Todd PK, Askari FK, and Fontana RJ
- Subjects
- Humans, RNA, Messenger, SARS-CoV-2, Vaccination adverse effects, COVID-19, Guillain-Barre Syndrome diagnosis, Guillain-Barre Syndrome etiology, Liver Transplantation adverse effects
- Published
- 2022
- Full Text
- View/download PDF
22. SRSF protein kinase 1 modulates RAN translation and suppresses CGG repeat toxicity.
- Author
-
Malik I, Tseng YJ, Wright SE, Zheng K, Ramaiyer P, Green KM, and Todd PK
- Subjects
- Animals, C9orf72 Protein genetics, C9orf72 Protein metabolism, Drosophila melanogaster genetics, Fragile X Mental Retardation Protein genetics, Fragile X Mental Retardation Protein metabolism, Trinucleotide Repeat Expansion, Amyotrophic Lateral Sclerosis genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism, Fragile X Syndrome genetics, Frontotemporal Dementia, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism
- Abstract
Transcribed CGG repeat expansions cause neurodegeneration in Fragile X-associated tremor/ataxia syndrome (FXTAS). CGG repeat RNAs sequester RNA-binding proteins (RBPs) into nuclear foci and undergo repeat-associated non-AUG (RAN) translation into toxic peptides. To identify proteins involved in these processes, we employed a CGG repeat RNA-tagging system to capture repeat-associated RBPs by mass spectrometry in mammalian cells. We identified several SR (serine/arginine-rich) proteins that interact selectively with CGG repeats basally and under cellular stress. These proteins modify toxicity in a Drosophila model of FXTAS. Pharmacologic inhibition of serine/arginine protein kinases (SRPKs), which alter SRSF protein phosphorylation, localization, and activity, directly inhibits RAN translation of CGG and GGGGCC repeats (associated with C9orf72 ALS/FTD) and triggers repeat RNA retention in the nucleus. Lowering SRPK expression suppressed toxicity in both FXTAS and C9orf72 ALS/FTD model flies, and SRPK inhibitors suppressed CGG repeat toxicity in rodent neurons. Together, these findings demonstrate roles for CGG repeat RNA binding proteins in RAN translation and repeat toxicity and support further evaluation of SRPK inhibitors in modulating RAN translation associated with repeat expansion disorders., (© 2021 The Authors Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
23. Two-Step Solid-State Synthesis of Ternary Nitride Materials.
- Author
-
Todd PK, Fallon MJ, Neilson JR, and Zakutayev A
- Abstract
Ternary nitride materials hold promise for many optical, electronic, and refractory applications; yet, their preparation via solid-state synthesis remains challenging. Often, high pressures or reactive gases are used to manipulate the effective chemical potential of nitrogen, yet these strategies require specialized equipment. Here, we report on a simple two-step synthesis using ion-exchange reactions that yield rocksalt-derived MgZrN
2 and Mg2 NbN3 , as well as layered MgMoN2 . All three compounds show almost temperature-independent and weak paramagnetic responses to an applied magnetic field at cryogenic temperatures, indicating phase-pure products. The key to synthesizing these ternary materials is an initial low-temperature step (300-450 °C) to promote Mg-M-N nucleation. The intermediates then are annealed (800-900 °C) to grow crystalline domains of the ternary product. Calorimetry experiments reveal that initial reaction temperatures are determined by phase transitions of reaction precursors, whereas heating directly to high temperatures results in decomposition. These two-step reactions provide a rational guide to material discovery of other bulk ternary nitrides., Competing Interests: The authors declare no competing financial interest., (© 2021 American Chemical Society.)- Published
- 2021
- Full Text
- View/download PDF
24. Mild Neurological Signs in FMR1 Premutation Women in an Unselected Community-Based Cohort.
- Author
-
Mailick MR, Hong J, Movaghar A, DaWalt L, Berry-Kravis EM, Brilliant MH, Boero J, Todd PK, and Hall D
- Subjects
- Ataxia genetics, Female, Humans, Tremor genetics, Trinucleotide Repeat Expansion, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Heterozygote
- Abstract
Background: Premutation-sized (55-200) CGG repeat expansions in the FMR1 gene cause fragile X-associated tremor/ataxia syndrome (FXTAS). Most studies of premutation carriers utilized reverse ascertainment to identify patients, leading to a selection bias for larger repeats. As shorter CGG premutation repeats are common in the population, understanding their impact on health outcomes has a potentially large public health footprint., Objective: The study's objective was to compare an unselected group of premutation carriers (n = 35, 55-101 CGG repeats) with matched controls (n = 61, 29-39 CGG repeats) with respect to FXTAS-type signs using structured neurological assessments., Methods: Three neurologists independently rated signs, using an adapted version of the FXTAS Rating Scale (Leehey MA, Berry-Kravis E, Goetz CG, et al. FMR1 CGG repeat length predicts motor dysfunction in premutation carriers. Neurology. 2008). This was a double-blind study, as genetic status (premutation vs. control) was known neither by the participants nor by any of the neurologists. Analyses controlled potentially confounding comorbid conditions in the electronic health record (eg, osteoarthritis and stroke) and probed the association of age with signs., Results: Although there was no overall difference between carriers and controls, among individuals without any potentially confounding comorbid diagnoses, there was a statistically significant age-associated elevation in FXTAS-type signs in premutation carriers compared to controls., Conclusions: Among those who do not have other comorbid diagnoses, women who have CGG repeats at the lower end of the premutation range may be at greater risk for ataxia and parkinsonism than their age peers, although their overall risk of developing such clinical features is low. This study should provide reassurance to those who share characteristics with the present cohort. © 2021 International Parkinson and Movement Disorder Society., (© 2021 International Parkinson and Movement Disorder Society.)
- Published
- 2021
- Full Text
- View/download PDF
25. Selectivity in Yttrium Manganese Oxide Synthesis via Local Chemical Potentials in Hyperdimensional Phase Space.
- Author
-
Todd PK, McDermott MJ, Rom CL, Corrao AA, Denney JJ, Dwaraknath SS, Khalifah PG, Persson KA, and Neilson JR
- Abstract
In sharp contrast to molecular synthesis, materials synthesis is generally presumed to lack selectivity. The few known methods of designing selectivity in solid-state reactions have limited scope, such as topotactic reactions or strain stabilization. This contribution describes a general approach for searching large chemical spaces to identify selective reactions. This novel approach explains the ability of a nominally "innocent" Na
2 CO3 precursor to enable the metathesis synthesis of single-phase Y2 Mn2 O7 : an outcome that was previously only accomplished at extreme pressures and which cannot be achieved with closely related precursors of Li2 CO3 and K2 CO3 under identical conditions. By calculating the required change in chemical potential across all possible reactant-product interfaces in an expanded chemical space including Y, Mn, O, alkali metals, and halogens, using thermodynamic parameters obtained from density functional theory calculations, we identify reactions that minimize the thermodynamic competition from intermediates. In this manner, only the Na-based intermediates minimize the distance in the hyperdimensional chemical potential space to Y2 Mn2 O7 , thus providing selective access to a phase which was previously thought to be metastable. Experimental evidence validating this mechanism for pathway-dependent selectivity is provided by intermediates identified from in situ synchrotron-based crystallographic analysis. This approach of calculating chemical potential distances in hyperdimensional compositional spaces provides a general method for designing selective solid-state syntheses that will be useful for gaining access to metastable phases and for identifying reaction pathways that can reduce the synthesis temperature, and cost, of technological materials.- Published
- 2021
- Full Text
- View/download PDF
26. Author Correction: Molecular mechanisms underlying nucleotide repeat expansion disorders.
- Author
-
Malik I, Kelley CP, Wang ET, and Todd PK
- Published
- 2021
- Full Text
- View/download PDF
27. Molecular mechanisms underlying nucleotide repeat expansion disorders.
- Author
-
Malik I, Kelley CP, Wang ET, and Todd PK
- Subjects
- Animals, Gene Silencing, Genomic Instability, Humans, Mutation, Neurodegenerative Diseases pathology, Neurodegenerative Diseases physiopathology, Organ Specificity, Protein Biosynthesis, R-Loop Structures, RNA chemistry, RNA metabolism, RNA-Binding Proteins metabolism, DNA Repeat Expansion genetics, Neurodegenerative Diseases genetics
- Abstract
The human genome contains over one million short tandem repeats. Expansion of a subset of these repeat tracts underlies over fifty human disorders, including common genetic causes of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (C9orf72), polyglutamine-associated ataxias and Huntington disease, myotonic dystrophy, and intellectual disability disorders such as Fragile X syndrome. In this Review, we discuss the four major mechanisms by which expansion of short tandem repeats causes disease: loss of function through transcription repression, RNA-mediated gain of function through gelation and sequestration of RNA-binding proteins, gain of function of canonically translated repeat-harbouring proteins, and repeat-associated non-AUG translation of toxic repeat peptides. Somatic repeat instability amplifies these mechanisms and influences both disease age of onset and tissue specificity of pathogenic features. We focus on the crosstalk between these disease mechanisms, and argue that they often synergize to drive pathogenesis. We also discuss the emerging native functions of repeat elements and how their dynamics might contribute to disease at a larger scale than currently appreciated. Lastly, we propose that lynchpins tying these disease mechanisms and native functions together offer promising therapeutic targets with potential shared applications across this class of human disorders., (© 2021. Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
28. The RNA helicase DHX36-G4R1 modulates C9orf72 GGGGCC hexanucleotide repeat-associated translation.
- Author
-
Tseng YJ, Sandwith SN, Green KM, Chambers AE, Krans A, Raimer HM, Sharlow ME, Reisinger MA, Richardson AE, Routh ED, Smaldino MA, Wang YH, Vaughn JP, Todd PK, and Smaldino PJ
- Subjects
- Amyotrophic Lateral Sclerosis enzymology, Amyotrophic Lateral Sclerosis genetics, C9orf72 Protein metabolism, Cell Line, Tumor, Frontotemporal Dementia enzymology, Frontotemporal Dementia genetics, Frontotemporal Dementia pathology, Humans, Protein Biosynthesis, Amyotrophic Lateral Sclerosis pathology, C9orf72 Protein genetics, DEAD-box RNA Helicases metabolism, DNA Repeat Expansion, G-Quadruplexes, RNA Helicases metabolism
- Abstract
GGGGCC (G
4 C2 ) hexanucleotide repeat expansions in the endosomal trafficking gene C9orf72 are the most common genetic cause of ALS and frontotemporal dementia. Repeat-associated non-AUG (RAN) translation of this expansion through near-cognate initiation codon usage and internal ribosomal entry generates toxic proteins that accumulate in patients' brains and contribute to disease pathogenesis. The helicase protein DEAH-box helicase 36 (DHX36-G4R1) plays active roles in RNA and DNA G-quadruplex (G4) resolution in cells. As G4 C2 repeats are known to form G4 structures in vitro, we sought to determine the impact of manipulating DHX36 expression on repeat transcription and RAN translation. Using a series of luciferase reporter assays both in cells and in vitro, we found that DHX36 depletion suppresses RAN translation in a repeat length-dependent manner, whereas overexpression of DHX36 enhances RAN translation from G4 C2 reporter RNAs. Moreover, upregulation of RAN translation that is typically triggered by integrated stress response activation is prevented by loss of DHX36. These results suggest that DHX36 is active in regulating G4 C2 repeat translation, providing potential implications for therapeutic development in nucleotide repeat expansion disorders., Competing Interests: Conflict of interest P. K. T. served as a paid consultant for Denali Therapeutics, holds a joint patent with Ionis Therapeutics, and receives publishing royalties from UpToDate. None of these are directly relevant to his role on this article, and none of these organizations have any role in the conception, preparation, or editing of this article. All other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
29. Human oncoprotein 5MP suppresses general and repeat-associated non-AUG translation via eIF3 by a common mechanism.
- Author
-
Singh CR, Glineburg MR, Moore C, Tani N, Jaiswal R, Zou Y, Aube E, Gillaspie S, Thornton M, Cecil A, Hilgers M, Takasu A, Asano I, Asano M, Escalante CR, Nakamura A, Todd PK, and Asano K
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Antigens, Differentiation metabolism, DNA-Binding Proteins chemistry, Drosophila metabolism, Drosophila Proteins metabolism, Eukaryotic Initiation Factor-2 metabolism, Eukaryotic Initiation Factor-3 chemistry, HEK293 Cells, Humans, Male, Models, Biological, Models, Molecular, Mutation genetics, Peptide Chain Initiation, Translational, Protein Binding, Protein Domains, Receptors, Immunologic metabolism, Codon, Initiator genetics, DNA-Binding Proteins metabolism, Eukaryotic Initiation Factor-3 metabolism, Protein Biosynthesis genetics, Trinucleotide Repeat Expansion genetics
- Abstract
eIF5-mimic protein (5MP) is a translational regulatory protein that binds the small ribosomal subunit and modulates its activity. 5MP is proposed to reprogram non-AUG translation rates for oncogenes in cancer, but its role in controlling non-AUG initiated synthesis of deleterious repeat-peptide products, such as FMRpolyG observed in fragile-X-associated tremor ataxia syndrome (FXTAS), is unknown. Here, we show that 5MP can suppress both general and repeat-associated non-AUG (RAN) translation by a common mechanism in a manner dependent on its interaction with eIF3. Essentially, 5MP displaces eIF5 through the eIF3c subunit within the preinitiation complex (PIC), thereby increasing the accuracy of initiation. In Drosophila, 5MP/Kra represses neuronal toxicity and enhances the lifespan in an FXTAS disease model. These results implicate 5MP in protecting cells from unwanted byproducts of non-AUG translation in neurodegeneration., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. Translational control in aging and neurodegeneration.
- Author
-
Skariah G and Todd PK
- Subjects
- Humans, RNA, Messenger metabolism, RNA, Ribosomal, Ribosomal Proteins metabolism, Ribosomes metabolism, Aging, Neurodegenerative Diseases genetics, Protein Biosynthesis genetics
- Abstract
Protein metabolism plays central roles in age-related decline and neurodegeneration. While a large body of research has explored age-related changes in protein degradation, alterations in the efficiency and fidelity of protein synthesis with aging are less well understood. Age-associated changes occur in both the protein synthetic machinery (ribosomal proteins and rRNA) and within regulatory factors controlling translation. At the same time, many of the interventions that prolong lifespan do so in part by pre-emptively decreasing protein synthesis rates to allow better harmonization to age-related declines in protein catabolism. Here we review the roles of translation regulation in aging, with a specific focus on factors implicated in age-related neurodegeneration. We discuss how emerging technologies such as ribosome profiling and superior mass spectrometric approaches are illuminating age-dependent mRNA-specific changes in translation rates across tissues to reveal a critical interplay between catabolic and anabolic pathways that likely contribute to functional decline. These new findings point to nodes in posttranscriptional gene regulation that both contribute to aging and offer targets for therapy. This article is categorized under: Translation > Translation Regulation Translation > Ribosome Biogenesis Translation > Translation Mechanisms., (© 2020 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
31. Enhanced detection of expanded repeat mRNA foci with hybridization chain reaction.
- Author
-
Glineburg MR, Zhang Y, Krans A, Tank EM, Barmada SJ, and Todd PK
- Subjects
- Amyotrophic Lateral Sclerosis metabolism, Amyotrophic Lateral Sclerosis pathology, C9orf72 Protein metabolism, Cells, Cultured, DNA Repeat Expansion physiology, Fibroblasts metabolism, Fibroblasts pathology, Frontotemporal Dementia metabolism, Frontotemporal Dementia pathology, Humans, RNA, Messenger metabolism, Amyotrophic Lateral Sclerosis genetics, C9orf72 Protein genetics, Frontotemporal Dementia genetics, In Situ Hybridization, Fluorescence methods, RNA, Messenger genetics
- Abstract
Transcribed nucleotide repeat expansions form detectable RNA foci in patient cells that contribute to disease pathogenesis. The most widely used method for detecting RNA foci, fluorescence in situ hybridization (FISH), is powerful but can suffer from issues related to signal above background. Here we developed a repeat-specific form of hybridization chain reaction (R-HCR) as an alternative method for detection of repeat RNA foci in two neurodegenerative disorders: C9orf72 associated ALS and frontotemporal dementia (C9 ALS/FTD) and Fragile X-associated tremor/ataxia syndrome. R-HCR to both G
4 C2 and CGG repeats exhibited comparable specificity but > 40 × sensitivity compared to FISH, with better detection of both nuclear and cytoplasmic foci in human C9 ALS/FTD fibroblasts, patient iPSC derived neurons, and patient brain samples. Using R-HCR, we observed that integrated stress response (ISR) activation significantly increased the number of endogenous G4 C2 repeat RNA foci and triggered their selective nuclear accumulation without evidence of stress granule co-localization in patient fibroblasts and patient derived neurons. These data suggest that R-HCR can be a useful tool for tracking the behavior of repeat expansion mRNA in C9 ALS/FTD and other repeat expansion disorders.- Published
- 2021
- Full Text
- View/download PDF
32. Neuropathology of FMR1 -premutation carriers presenting with dementia and neuropsychiatric symptoms.
- Author
-
Dijkstra AA, Haify SN, Verwey NA, Prins ND, van der Toorn EC, Rozemuller AJM, Bugiani M, den Dunnen WFA, Todd PK, Charlet-Berguerand N, Willemsen R, Hukema RK, and Hoozemans JJM
- Abstract
CGG repeat expansions within the premutation range (55-200) of the FMR1 gene can lead to Fragile X-associated tremor/ataxia syndrome and Fragile X-associated neuropsychiatric disorders. These CGG repeats are translated into a toxic polyglycine-containing protein, FMRpolyG. Pathology of Fragile X-associated tremor/ataxia syndrome and Fragile X-associated neuropsychiatric disorders comprises FMRpolyG- and p62-positive intranuclear inclusions. Diagnosing a FMR1 -premutation carrier remains challenging, as the clinical features overlap with other neurodegenerative diseases. Here, we describe two male cases with Fragile X-associated neuropsychiatric disorders-related symptoms and mild movement disturbances and novel pathological features that can attribute to the variable phenotype. Macroscopically, both donors did not show characteristic white matter lesions on MRI; however, vascular infarcts in cortical- and sub-cortical regions were identified. Immunohistochemistry analyses revealed a high number of FMRpolyG intranuclear inclusions throughout the brain, which were also positive for p62. Importantly, we identified a novel pathological vascular phenotype with inclusions present in pericytes and endothelial cells. Although these results need to be confirmed in more cases, we propose that these vascular lesions in the brain could contribute to the complex symptomology of FMR1 -premutation carriers. Overall, our report suggests that Fragile X-associated tremor/ataxia syndrome and Fragile X-associated neuropsychiatric disorders may present diverse clinical involvements resembling other types of dementia, and in the absence of genetic testing, FMRpolyG can be used post-mortem to identify premutation carriers., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
33. A repeating theme in amyotrophic lateral sclerosis genetics.
- Author
-
Todd PK
- Subjects
- Humans, Amyotrophic Lateral Sclerosis genetics
- Published
- 2020
- Full Text
- View/download PDF
34. Defect-Accommodating Intermediates Yield Selective Low-Temperature Synthesis of YMnO 3 Polymorphs.
- Author
-
Todd PK, Wustrow A, McAuliffe RD, McDermott MJ, Tran GT, McBride BC, Boeding ED, O'Nolan D, Liu CH, Dwaraknath SS, Chapman KW, Billinge SJL, Persson KA, Huq A, Veith GM, and Neilson JR
- Abstract
In the synthesis of complex oxides, solid-state metathesis provides low-temperature reactions where product selectivity can be achieved through simple changes in precursor composition. The influence of precursor structure, however, is less understood in solid-state synthesis. Here we present the ternary metathesis reaction (LiMnO
2 + YOCl → YMnO3 + LiCl) to target two yttrium manganese oxide products, hexagonal and orthorhombic YMnO3 , when starting from three different LiMnO2 precursors. Using temperature-dependent synchrotron X-ray and neutron diffraction, we identify the relevant intermediates and temperature regimes of reactions along the pathway to YMnO3 . Manganese-containing intermediates undergo a charge disproportionation into a reduced Mn(II,III) tetragonal spinel and oxidized Mn(III,IV) cubic spinel, which lead to hexagonal and orthorhombic YMnO3 , respectively. Density functional theory calculations confirm that the presence of Mn(IV) caused by a small concentration of cation vacancies (∼2.2%) in YMnO3 stabilizes the orthorhombic polymorph over the hexagonal. Reactions over the course of 2 weeks yield o-YMnO3 as the majority product at temperatures below 600 °C, which supports an equilibration of cation defects over time. Controlling the composition and structure of these defect-accommodating intermediates provides new strategies for selective synthesis of complex oxides at low temperatures.- Published
- 2020
- Full Text
- View/download PDF
35. Catalytic behavior of hexaphenyldisiloxane in the synthesis of pyrite FeS 2 .
- Author
-
Todd PK, Martinolich AJ, and Neilson JR
- Abstract
Functional small molecules afford opportunities to direct solid-state inorganic reactions at low temperatures. Here, we use catalytic amounts of organosilicon molecules to influence the metathesis reaction: FeCl2 + Na2S2 → 2NaCl + FeS2. Specifically, hexaphenyldisiloxane ((C6H5)6Si2O) is shown to increase pyrite yields in metathesis reactions performed at 150 °C. In situ synchrotron X-ray diffraction (SXRD) paired with differential scanning calorimetry (DSC) reveals that diffusion-limited intermediates are circumvented in the presence of (C6H5)6Si2O. Control reactions suggest that the observed change in the reaction pathway is imparted by the Si-O functional group. 1H NMR supports catalytic behavior, as (C6H5)6Si2O is unchanged ex post facto. Taken together, we hypothesize that the polar Si-O functional group coordinates to iron chloride species when NaCl and Na2S4 form, forming an unidentified, transient intermediate. Further exploration of targeted small molecules in these metathesis reaction provides new strategies in controlling inorganic materials synthesis at low-temperatures.
- Published
- 2020
- Full Text
- View/download PDF
36. The carboxyl termini of RAN translated GGGGCC nucleotide repeat expansions modulate toxicity in models of ALS/FTD.
- Author
-
He F, Flores BN, Krans A, Frazer M, Natla S, Niraula S, Adefioye O, Barmada SJ, and Todd PK
- Subjects
- Animals, Dipeptides, Disease Models, Animal, Drosophila, Rats, Rats, Long-Evans, Amyotrophic Lateral Sclerosis genetics, C9orf72 Protein genetics, DNA Repeat Expansion, Frontotemporal Dementia genetics, Protein Biosynthesis genetics, Reading Frames genetics
- Abstract
An intronic hexanucleotide repeat expansion in C9ORF72 causes familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This repeat is thought to elicit toxicity through RNA mediated protein sequestration and repeat-associated non-AUG (RAN) translation of dipeptide repeat proteins (DPRs). We generated a series of transgenic Drosophila models expressing GGGGCC (G
4 C2 ) repeats either inside of an artificial intron within a GFP reporter or within the 5' untranslated region (UTR) of GFP placed in different downstream reading frames. Expression of 484 intronic repeats elicited minimal alterations in eye morphology, viability, longevity, or larval crawling but did trigger RNA foci formation, consistent with prior reports. In contrast, insertion of repeats into the 5' UTR elicited differential toxicity that was dependent on the reading frame of GFP relative to the repeat. Greater toxicity correlated with a short and unstructured carboxyl terminus (C-terminus) in the glycine-arginine (GR) RAN protein reading frame. This change in C-terminal sequence triggered nuclear accumulation of all three RAN DPRs. A similar differential toxicity and dependence on the GR C-terminus was observed when repeats were expressed in rodent neurons. The presence of the native C-termini across all three reading frames was partly protective. Taken together, these findings suggest that C-terminal sequences outside of the repeat region may alter the behavior and toxicity of dipeptide repeat proteins derived from GGGGCC repeats.- Published
- 2020
- Full Text
- View/download PDF
37. A thermal-gradient approach to variable-temperature measurements resolved in space.
- Author
-
O'Nolan D, Huang G, Kamm GE, Grenier A, Liu CH, Todd PK, Wustrow A, Thinh Tran G, Montiel D, Neilson JR, Billinge SJL, Chupas PJ, Thornton KS, and Chapman KW
- Abstract
Temperature is a ubiquitous environmental variable used to explore materials structure, properties and reactivity. This article reports a new paradigm for variable-temperature measurements that varies the temperature continuously across a sample such that temperature is measured as a function of sample position and not time. The gradient approach offers advantages over conventional variable-temperature studies, in which temperature is scanned during a series measurement, in that it improves the efficiency with which a series of temperatures can be probed and it allows the sample evolution at multiple temperatures to be measured in parallel to resolve kinetic and thermodynamic effects. Applied to treat samples at a continuum of tem-peratures prior to measurements at ambient temperature, the gradient approach enables parametric studies of recovered systems, eliminating temperature-dependent structural and chemical variations to simplify interpretation of the data. The implementation of spatially resolved variable-temperature measurements presented here is based on a gradient-heater design that uses a 3D-printed ceramic template to guide the variable pitch of the wire in a resistively heated wire-wound heater element. The configuration of the gradient heater was refined on the basis of thermal modelling. Applications of the gradient heater to quantify thermal-expansion behaviour, to map metastable polymorphs recovered to ambient temperature, and to monitor the time- and temperature-dependent phase evolution in a complex solid-state reaction are demonstrated., (© International Union of Crystallography 2020.)
- Published
- 2020
- Full Text
- View/download PDF
38. A native function for RAN translation and CGG repeats in regulating fragile X protein synthesis.
- Author
-
Rodriguez CM, Wright SE, Kearse MG, Haenfler JM, Flores BN, Liu Y, Ifrim MF, Glineburg MR, Krans A, Jafar-Nejad P, Sutton MA, Bassell GJ, Parent JM, Rigo F, Barmada SJ, and Todd PK
- Subjects
- Animals, Cell Line, Cell Survival genetics, Female, Fragile X Mental Retardation Protein biosynthesis, Induced Pluripotent Stem Cells, Male, Mice, Neurons metabolism, Oligonucleotides, Antisense pharmacology, Protein Biosynthesis, Rats, Rats, Long-Evans, Rats, Sprague-Dawley, Receptor, Metabotropic Glutamate 5 biosynthesis, Receptor, Metabotropic Glutamate 5 genetics, DNA Repeat Expansion genetics, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, Trinucleotide Repeats genetics
- Abstract
Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.
- Published
- 2020
- Full Text
- View/download PDF
39. Genetic testing utilization for patients with neurologic disease and the limitations of claims data.
- Author
-
Mackenzie SJ, Lin CC, Todd PK, Burke JF, and Callaghan BC
- Abstract
Objective: To determine the utilization of genetic testing in patients seen by a neurologist within a large private insurance population., Methods: Using the Optum health care claims database, we identified a cross-sectional cohort of patients who had been evaluated by a neurologist no more than 30 days before initial genetic testing. Within this group, we then categorized genetic testing between 2014 and 2016 on the basis of the Current Procedural Terminology (CPT) codes related to molecular and genetic testing. We also evaluated the International Classification of Disease Version 9 Clinical Code Classifications (ICD-9 CCS) associated with testing., Results: From 2014 to 2016, a total of 45,014 claims were placed for 29,951 patients who had been evaluated by a neurologist within the preceding 30 days. Of these, 29,926 (66.5%) were associated with codes that were too nonspecific to infer what test was actually performed. Among those claims where the test was clearly identifiable, 7,307 (16.2%) were likely obtained for purposes of neurologic diagnosis, whereas the remainder (17.2%) was obtained for non-neurological purposes. An additional 3,793 claims (8.4%) wherein the test ordered could not be clearly identified were associated with a neurology-related ICD-9 CCS., Conclusions: Accurate assessment of genetic testing utilization using claims data is not possible given the high prevalence of nonspecific codes. Reducing the ambiguity surrounding the CPT codes and the actual testing performed will become even more important as more genetic tests become available., (Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.)
- Published
- 2020
- Full Text
- View/download PDF
40. High-throughput screening yields several small-molecule inhibitors of repeat-associated non-AUG translation.
- Author
-
Green KM, Sheth UJ, Flores BN, Wright SE, Sutter AB, Kearse MG, Barmada SJ, Ivanova MI, and Todd PK
- Subjects
- Amyotrophic Lateral Sclerosis drug therapy, Animals, Ataxia drug therapy, Azepines pharmacology, Azepines therapeutic use, Cells, Cultured, Circular Dichroism, DNA Repeat Expansion drug effects, DNA Repeat Expansion genetics, Drug Evaluation, Preclinical, Fragile X Syndrome drug therapy, HEK293 Cells, Humans, Neurodegenerative Diseases genetics, Propidium pharmacology, Propidium therapeutic use, Pyrimidines pharmacology, Pyrimidines therapeutic use, Quinazolines pharmacology, Quinazolines therapeutic use, Rats, Tremor drug therapy, Trinucleotide Repeat Expansion drug effects, Amyotrophic Lateral Sclerosis genetics, Ataxia genetics, Fragile X Syndrome genetics, Tremor genetics, Trinucleotide Repeat Expansion genetics
- Abstract
Repeat-associated non-AUG (RAN) translation is a noncanonical translation initiation event that occurs at nucleotide-repeat expansion mutations that are associated with several neurodegenerative diseases, including fragile X-associated tremor ataxia syndrome (FXTAS), ALS, and frontotemporal dementia (FTD). Translation of expanded repeats produces toxic proteins that accumulate in human brains and contribute to disease pathogenesis. Consequently, RAN translation constitutes a potentially important therapeutic target for managing multiple neurodegenerative disorders. Here, we adapted a previously developed RAN translation assay to a high-throughput format to screen 3,253 bioactive compounds for inhibition of RAN translation of expanded CGG repeats associated with FXTAS. We identified five diverse small molecules that dose-dependently inhibited CGG RAN translation, while relatively sparing canonical translation. All five compounds also inhibited RAN translation of expanded GGGGCC repeats associated with ALS and FTD. Using CD and native gel analyses, we found evidence that three of these compounds, BIX01294, CP-31398, and propidium iodide, bind directly to the repeat RNAs. These findings provide proof-of-principle supporting the development of selective small-molecule RAN translation inhibitors that act across multiple disease-causing repeats.
- Published
- 2019
- Full Text
- View/download PDF
41. Yttrium Manganese Oxide Phase Stability and Selectivity Using Lithium Carbonate Assisted Metathesis Reactions.
- Author
-
Todd PK, Smith AMM, and Neilson JR
- Abstract
In solid-state chemistry, stable phases are often missed if their synthesis is impractical, such as when decomposition or a polymorphic transition occurs at relatively low temperature. In the preparation of complex oxides, reaction temperatures commonly exceed 1000 °C with little to no control of the reaction pathway. Thus, a prerequisite for exploring the synthesis of complex oxides is to identify reactions with intermediates that are kinetically competent at low temperatures, as provided by assisted metathesis reactions. Here, we study the assisted metathesis reaction Mn
2 O3 + 2.2YCl3 ·6H2 O + 3Li2 CO3 → 2YMnO3 + 5.8LiCl + 0.2LiYCl4 + 3CO2 using in situ synchrotron X-ray diffraction. By changing the atmosphere, oxygen vs inert gas, the reaction product changes from the overoxidized perovskite YMnO3+δ to the hexagonal YMnO3 polymorph at the reaction temperature of 850 °C, respectively. Analysis of the reaction pathways reveals two parallel reaction pathways in forming YMnO3 phases: (1) the slow reaction of metal oxides in a LiCl flux (Y2 O3 + Mn2 O3 [Formula: see text] 2YMnO3 ) and (2) the fast reaction from ternary intermediates (YOCl + LiMnO2 → LiCl + YMnO3 ). Control reactions reveal that both proposed pathways in isolation result in product formation, but the direct preparation of ternary intermediates (YOCl + LiMnO2 → LiCl + YMnO3 ) occurs at lower temperatures (500 °C) and shorter times (<24 h) and forms nominally stoichiometric orthorhombic YMnO3 . These ternary intermediates react at a faster rate than the slow stepwise oxygenation of yttrium chloride to Y2 O3 (YCl3 → YOCl → Y3 O4 Cl → Y2 O3 ), which is relatively inert. These results support a kinetically controlled reaction pathway to form YMnO3 phases in assisted metathesis reactions with phase selectivity achievable through changes to reaction atmosphere.- Published
- 2019
- Full Text
- View/download PDF
42. Neuropathology of RAN translation proteins in fragile X-associated tremor/ataxia syndrome.
- Author
-
Krans A, Skariah G, Zhang Y, Bayly B, and Todd PK
- Subjects
- Fragile X Mental Retardation Protein metabolism, Gene Expression, HEK293 Cells, Humans, Intranuclear Inclusion Bodies metabolism, Intranuclear Inclusion Bodies pathology, RNA-Binding Proteins metabolism, Sequestosome-1 Protein metabolism, Trinucleotide Repeat Expansion, Ubiquitin metabolism, Ataxia metabolism, Ataxia pathology, Brain metabolism, Brain pathology, Fragile X Syndrome metabolism, Fragile X Syndrome pathology, Neurons metabolism, Neurons pathology, Tremor metabolism, Tremor pathology
- Abstract
CGG repeat expansions in FMR1 cause the neurodegenerative disorder Fragile X-associated Tremor/Ataxia Syndrome (FXTAS). Ubiquitinated neuronal intranuclear inclusions (NIIs) are the neuropathological hallmark of FXTAS. Both sense strand derived CGG repeats and antisense strand derived CCG repeats support non-AUG initiated (RAN) translation of homopolymeric proteins in potentially 6 different reading frames. However, the relative abundance of these proteins in FXTAS brains and their co-localization with each other and NIIs is lacking. Here we describe rater-blinded assessment of immunohistochemical and immunofluorescence staining with newly generated antibodies to different CGG RAN translation products in FXTAS and control brains as well as co-staining with ubiquitin, p62/SQSTM1, and ubiquilin 2. We find that both FMRpolyG and a second CGG repeat derived RAN translation product, FMRpolyA, accumulate in aggregates in FXTAS brains. FMRpolyG is a near-obligate component of both ubiquitin-positive and p62-positive NIIs in FXTAS, with occurrence of aggregates in 20% of all hippocampal neurons and > 90% of all inclusions. A subset of these inclusions also stain positive for the ALS/FTD associated protein ubiquilin 2. Ubiquitinated inclusions and FMRpolyG+ aggregates are rarer in cortex and cerebellum. Intriguingly, FMRpolyG staining is also visible in control neuronal nuclei. In contrast to FMRpolyG, staining for FMRpolyA and CCG antisense derived RAN translation products were less abundant and less frequent components of ubiquitinated inclusions. In conclusion, RAN translated FMRpolyG is a common component of ubiquitin and p62 positive inclusions in FXTAS patient brains.
- Published
- 2019
- Full Text
- View/download PDF
43. Fragile X-associated tremor ataxia syndrome with co-occurrent progressive supranuclear palsy-like neuropathology.
- Author
-
Sacino AN, Prokop S, Walsh MA, Adamson J, Subramony SH, Krans A, Todd PK, Giasson BI, and Yachnis AT
- Subjects
- Aged, Ataxia complications, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome complications, Humans, Intranuclear Inclusion Bodies pathology, Male, Neuroglia pathology, Neurons pathology, Supranuclear Palsy, Progressive complications, Tremor complications, Ataxia pathology, Brain pathology, Fragile X Syndrome pathology, Supranuclear Palsy, Progressive pathology, Tremor pathology
- Abstract
Co-occurrence of multiple neuropathologic changes is a common phenomenon, most prominently seen in Alzheimer's disease (AD) and Parkinson's disease (PD), complicating clinical diagnosis and patient management. Reports of co-occurring pathological processes are emerging in the group of genetically defined repeat-associated non-AUG (RAN)-translation related diseases. Here we report a case of Fragile X-associated tremor-ataxia syndrome (FXTAS) with widespread and abundant nuclear inclusions of the RAN-translation related FMRpolyG-peptide. In addition, we describe prominent neuronal and glial tau pathology representing changes seen in progressive supranuclear palsy (PSP). The highest abundance of the respective pathological changes was seen in distinct brain regions indicating an incidental, rather than causal correlation.
- Published
- 2019
- Full Text
- View/download PDF
44. New pathologic mechanisms in nucleotide repeat expansion disorders.
- Author
-
Rodriguez CM and Todd PK
- Subjects
- Humans, Neurodegenerative Diseases genetics, Brain pathology, DNA Repeat Expansion, Microsatellite Repeats, Neurodegenerative Diseases pathology
- Abstract
Tandem microsatellite repeats are common throughout the human genome and intrinsically unstable, exhibiting expansions and contractions both somatically and across generations. Instability in a small subset of these repeats are currently linked to human disease, although recent findings suggest more disease-causing repeats await discovery. These nucleotide repeat expansion disorders (NREDs) primarily affect the nervous system and commonly lead to neurodegeneration through toxic protein gain-of-function, protein loss-of-function, and toxic RNA gain-of-function mechanisms. However, the lines between these categories have blurred with recent findings of unconventional Repeat Associated Non-AUG (RAN) translation from putatively non-coding regions of the genome. Here we review two emerging topics in NREDs: 1) The mechanisms by which RAN translation occurs and its role in disease pathogenesis and 2) How nucleotide repeats as RNA and translated proteins influence liquid-liquid phase separation, membraneless organelle dynamics, and nucleocytoplasmic transport. We examine these topics with a particular eye on two repeats: the CGG repeat expansion responsible for Fragile X syndrome and Fragile X-associated Tremor Ataxia Syndrome (FXTAS) and the intronic GGGGCC repeat expansion in C9orf72, the most common inherited cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Our thesis is that these emerging disease mechanisms can inform a broader understanding of the native roles of microsatellites in cellular function and that aberrations in these native processes provide clues to novel therapeutic strategies for these currently untreatable disorders., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
45. DDX3X and specific initiation factors modulate FMR1 repeat-associated non-AUG-initiated translation.
- Author
-
Linsalata AE, He F, Malik AM, Glineburg MR, Green KM, Natla S, Flores BN, Krans A, Archbold HC, Fedak SJ, Barmada SJ, and Todd PK
- Subjects
- Animals, Ataxia genetics, Cells, Cultured, DEAD-box RNA Helicases genetics, Drosophila Proteins genetics, Drosophila melanogaster, Eukaryotic Initiation Factors genetics, Female, Fragile X Mental Retardation Protein genetics, Fragile X Syndrome genetics, HEK293 Cells, HeLa Cells, Humans, Immunoprecipitation, Male, Phenotype, Reverse Transcriptase Polymerase Chain Reaction, Tremor genetics, Ataxia metabolism, DEAD-box RNA Helicases metabolism, Drosophila Proteins metabolism, Eukaryotic Initiation Factors metabolism, Fragile X Mental Retardation Protein metabolism, Fragile X Syndrome metabolism, Tremor metabolism
- Abstract
A CGG trinucleotide repeat expansion in the 5' UTR of FMR1 causes the neurodegenerative disorder Fragile X-associated tremor/ataxia syndrome (FXTAS). This repeat supports a non-canonical mode of protein synthesis known as repeat-associated, non-AUG (RAN) translation. The mechanism underlying RAN translation at CGG repeats remains unclear. To identify modifiers of RAN translation and potential therapeutic targets, we performed a candidate-based screen of eukaryotic initiation factors and RNA helicases in cell-based assays and a Drosophila melanogaster model of FXTAS. We identified multiple modifiers of toxicity and RAN translation from an expanded CGG repeat in the context of the FMR1 5'UTR. These include the DEAD-box RNA helicase belle/DDX3X, the helicase accessory factors EIF4B/4H, and the start codon selectivity factors EIF1 and EIF5. Disrupting belle/DDX3X selectively inhibited FMR1 RAN translation in Drosophila in vivo and cultured human cells, and mitigated repeat-induced toxicity in Drosophila and primary rodent neurons. These findings implicate RNA secondary structure and start codon fidelity as critical elements mediating FMR1 RAN translation and identify potential targets for treating repeat-associated neurodegeneration., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
46. Correction to "Selective Formation of Yttrium Manganese Oxides through Kinetically Competent Assisted Metathesis Reactions".
- Author
-
Todd PK and Neilson JR
- Published
- 2019
- Full Text
- View/download PDF
47. Ribosome queuing enables non-AUG translation to be resistant to multiple protein synthesis inhibitors.
- Author
-
Kearse MG, Goldman DH, Choi J, Nwaezeapu C, Liang D, Green KM, Goldstrohm AC, Todd PK, Green R, and Wilusz JE
- Subjects
- Cycloheximide pharmacology, Eukaryotic Initiation Factor-4G genetics, Gene Expression Regulation drug effects, Genes, Reporter genetics, HEK293 Cells, HeLa Cells, Humans, Protein Synthesis Inhibitors pharmacology, Codon, Initiator genetics, Drug Resistance, Multiple genetics, Ribosomes genetics, Transcription Elongation, Genetic drug effects
- Abstract
Aberrant translation initiation at non-AUG start codons is associated with multiple cancers and neurodegenerative diseases. Nevertheless, how non-AUG translation may be regulated differently from canonical translation is poorly understood. Here, we used start codon-specific reporters and ribosome profiling to characterize how translation from non-AUG start codons responds to protein synthesis inhibitors in human cells. These analyses surprisingly revealed that translation of multiple non-AUG-encoded reporters and the endogenous GUG-encoded DAP5 ( eIF4G2 / p97 ) mRNA is resistant to cycloheximide (CHX), a translation inhibitor that severely slows but does not completely abrogate elongation. Our data suggest that slowly elongating ribosomes can lead to queuing/stacking of scanning preinitiation complexes (PICs), preferentially enhancing recognition of weak non-AUG start codons. Consistent with this model, limiting PIC formation or scanning sensitizes non-AUG translation to CHX. We further found that non-AUG translation is resistant to other inhibitors that target ribosomes within the coding sequence but not those targeting newly initiated ribosomes. Together, these data indicate that ribosome queuing enables mRNAs with poor initiation context-namely, those with non-AUG start codons-to be resistant to pharmacological translation inhibitors at concentrations that robustly inhibit global translation., (© 2019 Kearse et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
48. Translation of upstream open reading frames in a model of neuronal differentiation.
- Author
-
Rodriguez CM, Chun SY, Mills RE, and Todd PK
- Subjects
- Algorithms, Cell Line, Tumor, Gene Expression Regulation, Humans, Models, Biological, Neurons cytology, Ribosomes metabolism, Cell Differentiation genetics, Neurons metabolism, Open Reading Frames, Protein Biosynthesis
- Abstract
Background: Upstream open reading frames (uORFs) initiate translation within mRNA 5' leaders, and have the potential to alter main coding sequence (CDS) translation on transcripts in which they reside. Ribosome profiling (RP) studies suggest that translating ribosomes are pervasive within 5' leaders across model systems. However, the significance of this observation remains unclear. To explore a role for uORF usage in a model of neuronal differentiation, we performed RP on undifferentiated and differentiated human neuroblastoma cells., Results: Using a spectral coherence algorithm (SPECtre), we identify 4954 consistently translated uORFs across 31% of all neuroblastoma transcripts. These uORFs predominantly utilize non-AUG initiation codons and exhibit translational efficiencies (TE) comparable to annotated coding regions. On a population basis, the global impact of both AUG and non-AUG initiated uORFs on basal CDS translation were small, even when analysis is limited to conserved and consistently translated uORFs. However, uORFs did alter the translation of a subset of genes, including the Diamond-Blackfan Anemia associated ribosomal gene RPS24. With retinoic acid induced differentiation, we observed an overall positive correlation in translational shifts between uORF/CDS pairs. However, CDSs downstream of uORFs show smaller shifts in TE with differentiation relative to CDSs without a predicted uORF, suggesting that uORF translation buffers cell state dependent fluctuations in CDS translation., Conclusion: This work provides insights into the dynamic relationships and potential regulatory functions of uORF/CDS pairs in a model of neuronal differentiation.
- Published
- 2019
- Full Text
- View/download PDF
49. Selective Formation of Yttrium Manganese Oxides through Kinetically Competent Assisted Metathesis Reactions.
- Author
-
Todd PK and Neilson JR
- Abstract
The synthesis of complex oxides requires high temperatures to overcome barriers imparted by solid-state diffusion; as such, reactions typically yield the most stable polymorph for a given composition. To synthesize new or metastable complex oxides, kinetically competent reactions with lower initial energy barriers must be devised to control the reaction pathway and resulting products. This contribution details the selective synthesis of different yttrium manganese oxides through assisted metathesis reactions between Mn
2 O3 , YCl3 , and A2 CO3 under flowing oxygen; where A = Li, Na, K. With lithium carbonate, the orthorhombic perovskite o-YMnO3 (o-YMnO3+δ ) forms over the temperature range of 550-850 °C. With sodium carbonate, the pyrochlore Y2 Mn2 O7 forms at 650 °C. No apparent selectivity is observed with K2 CO3 , and all alkalis yields hexagonal YMnO3 at T > 950 °C. The alkali species modify the reaction pathway and thus impart kinetic control in the formation of both phases.- Published
- 2019
- Full Text
- View/download PDF
50. Targeted Reactivation of FMR1 Transcription in Fragile X Syndrome Embryonic Stem Cells.
- Author
-
Haenfler JM, Skariah G, Rodriguez CM, Monteiro da Rocha A, Parent JM, Smith GD, and Todd PK
- Abstract
Fragile X Syndrome (FXS) is the most common inherited cause of intellectual disability and autism. It results from expansion of a CGG nucleotide repeat in the 5' untranslated region (UTR) of FMR1 . Large expansions elicit repeat and promoter hyper-methylation, heterochromatin formation, FMR1 transcriptional silencing and loss of the Fragile X protein, FMRP. Efforts aimed at correcting the sequelae resultant from FMRP loss have thus far proven insufficient, perhaps because of FMRP's pleiotropic functions. As the repeats do not disrupt the FMRP coding sequence, reactivation of endogenous FMR1 gene expression could correct the proximal event in FXS pathogenesis. Here we utilize the Clustered Regularly Interspaced Palindromic Repeats/deficient CRISPR associated protein 9 (CRISPR/dCas9) system to selectively re-activate transcription from the silenced FMR1 locus. Fusion of the transcriptional activator VP192 to dCas9 robustly enhances FMR1 transcription and increases FMRP levels when targeted directly to the CGG repeat in human cells. Using a previously uncharacterized FXS human embryonic stem cell (hESC) line which acquires transcriptional silencing with serial passaging, we achieved locus-specific transcriptional re-activation of FMR1 messenger RNA (mRNA) expression despite promoter and repeat methylation. However, these changes at the transcript level were not coupled with a significant elevation in FMRP protein expression in FXS cells. These studies demonstrate that directing a transcriptional activator to CGG repeats is sufficient to selectively reactivate FMR1 mRNA expression in Fragile X patient stem cells.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.