51
|
Sareen D, O'Rourke JG, Meera P, Muhammad AKMG, Grant S, Simpkinson M, Bell S, Carmona S, Ornelas L, Sahabian A, Gendron T, Petrucelli L, Baughn M, Ravits J, Harms MB, Rigo F, Bennett CF, Otis TS, Svendsen CN, Baloh RH. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci Transl Med 2014; 5:208ra149. [PMID: 24154603 DOI: 10.1126/scitranslmed.3007529] [Citation(s) in RCA: 525] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative condition characterized by loss of motor neurons in the brain and spinal cord. Expansions of a hexanucleotide repeat (GGGGCC) in the noncoding region of the C9ORF72 gene are the most common cause of the familial form of ALS (C9-ALS), as well as frontotemporal lobar degeneration and other neurological diseases. How the repeat expansion causes disease remains unclear, with both loss of function (haploinsufficiency) and gain of function (either toxic RNA or protein products) proposed. We report a cellular model of C9-ALS with motor neurons differentiated from induced pluripotent stem cells (iPSCs) derived from ALS patients carrying the C9ORF72 repeat expansion. No significant loss of C9ORF72 expression was observed, and knockdown of the transcript was not toxic to cultured human motor neurons. Transcription of the repeat was increased, leading to accumulation of GGGGCC repeat-containing RNA foci selectively in C9-ALS iPSC-derived motor neurons. Repeat-containing RNA foci colocalized with hnRNPA1 and Pur-α, suggesting that they may be able to alter RNA metabolism. C9-ALS motor neurons showed altered expression of genes involved in membrane excitability including DPP6, and demonstrated a diminished capacity to fire continuous spikes upon depolarization compared to control motor neurons. Antisense oligonucleotides targeting the C9ORF72 transcript suppressed RNA foci formation and reversed gene expression alterations in C9-ALS motor neurons. These data show that patient-derived motor neurons can be used to delineate pathogenic events in ALS.
Collapse
Affiliation(s)
- Dhruv Sareen
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Abstract
Our understanding of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease, is expanding rapidly as its genetic causes are uncovered. The pace of new gene discovery over the last 5 years has accelerated, providing new insights into the pathogenesis of disease and highlighting biological pathways as targets for therapeutic development. This article reviews our current understanding of the heritability of ALS and provides an overview of each of the major ALS genes, highlighting their phenotypic characteristics and frequencies as a guide for clinicians evaluating patients with ALS.
Collapse
Affiliation(s)
- Matthew B Harms
- Neuromuscular Division, Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| | | |
Collapse
|
53
|
Abstract
CNG repeats (where N denotes one of the four natural nucleotides) are abundant in the human genome. Their tendency to undergo expansion can lead to hereditary diseases known as TREDs (trinucleotide repeat expansion disorders). The toxic factor can be protein, if the abnormal gene is expressed, or the gene transcript, or both. The gene transcripts have attracted much attention in the biomedical community, but their molecular structures have only recently been investigated. Model RNA molecules comprising CNG repeats fold into long hairpins whose stems generally conform to an A-type helix, in which the non-canonical N-N pairs are flanked by C-G and G-C pairs. Each homobasic pair is accommodated in the helical context in a unique manner, with consequences for the local helical parameters, solvent structure, electrostatic potential and potential to interact with ligands. The detailed three-dimensional profiles of RNA CNG repeats can be used in screening of compound libraries for potential therapeutics and in structure-based drug design. Here is a brief survey of the CNG structures published to date.
Collapse
Affiliation(s)
- Agnieszka Kiliszek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Wojciech Rypniewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
54
|
Antisense oligonucleotide therapy for the treatment of C9ORF72 ALS/FTD diseases. Mol Neurobiol 2014; 50:721-32. [PMID: 24809691 DOI: 10.1007/s12035-014-8724-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 04/28/2014] [Indexed: 10/25/2022]
Abstract
Motor neuron disorders, and particularly amyotrophic lateral sclerosis (ALS), are fatal diseases that are due to the loss of motor neurons in the brain and spinal cord, with progressive paralysis and premature death. It has been recently shown that the most frequent genetic cause of ALS, frontotemporal dementia (FTD), and other neurological diseases is the expansion of a hexanucleotide repeat (GGGGCC) in the non-coding region of the C9ORF72 gene. The pathogenic mechanisms that produce cell death in the presence of this expansion are still unclear. One of the most likely hypotheses seems to be the gain-of-function that is achieved through the production of toxic RNA (able to sequester RNA-binding protein) and/or toxic proteins. In recent works, different authors have reported that antisense oligonucleotides complementary to the C9ORF72 RNA transcript sequence were able to significantly reduce RNA foci generated by the expanded RNA, in affected cells. Here, we summarize the recent findings that support the idea that the buildup of "toxic" RNA containing the GGGGCC repeat contributes to the death of motor neurons in ALS and also suggest that the use of antisense oligonucleotides targeting this transcript is a promising strategy for treating ALS/frontotemporal lobe dementia (FTLD) patients with the C9ORF72 repeat expansion. These data are particularly important, given the state of the art antisense technology, and they allow researchers to believe that a clinical application of these discoveries will be possible soon.
Collapse
|
55
|
Mohan A, Goodwin M, Swanson MS. RNA-protein interactions in unstable microsatellite diseases. Brain Res 2014; 1584:3-14. [PMID: 24709120 DOI: 10.1016/j.brainres.2014.03.039] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/25/2014] [Accepted: 03/26/2014] [Indexed: 12/14/2022]
Abstract
A novel RNA-mediated disease mechanism has emerged from studies on dominantly inherited neurological disorders caused by unstable microsatellite expansions in non-coding regions of the genome. These non-coding tandem repeat expansions trigger the production of unusual RNAs that gain a toxic function, which involves the formation of RNA repeat structures that interact with, and alter the activities of, various factors required for normal RNA processing as well as additional cellular functions. In this review, we explore the deleterious effects of toxic RNA expression and discuss the various model systems currently available for studying RNA gain-of-function in neurologic diseases. Common themes, including bidirectional transcription and repeat-associated non-ATG (RAN) translation, have recently emerged from expansion disease studies. These and other discoveries have highlighted the need for further investigations designed to provide the additional mechanistic insights essential for future therapeutic development.
Collapse
Affiliation(s)
- Apoorva Mohan
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Cancer Genetics Research Complex, 2033 Mowry Road, Gainesville, FL 32610-3610, USA
| | - Marianne Goodwin
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Cancer Genetics Research Complex, 2033 Mowry Road, Gainesville, FL 32610-3610, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Cancer Genetics Research Complex, 2033 Mowry Road, Gainesville, FL 32610-3610, USA.
| |
Collapse
|
56
|
|
57
|
Gomes C, Merianda TT, Lee SJ, Yoo S, Twiss JL. Molecular determinants of the axonal mRNA transcriptome. Dev Neurobiol 2014; 74:218-32. [PMID: 23959706 PMCID: PMC3933445 DOI: 10.1002/dneu.22123] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/05/2013] [Accepted: 08/13/2013] [Indexed: 12/12/2022]
Abstract
Axonal protein synthesis has been shown to play a role in developmental and regenerative growth, as well as in cell body responses to axotomy. Recent studies have begun to identify the protein products that contribute to these autonomous responses of axons. In the peripheral nervous system, intra-axonal protein synthesis has been implicated in the localized in vivo responses to neuropathic stimuli, and there is emerging evidence for protein synthesis in CNS axons in vivo. Despite that hundreds of mRNAs have now been shown to localize into the axonal compartment, knowledge of what RNA binding proteins are responsible for this is quite limited. Here, we review the current state of knowledge of RNA transport mechanisms and highlight recently uncovered mechanisms for dynamically altering the axonal transcriptome. Both changes in the levels or activities of components of the RNA transport apparatus and alterations in transcription of transported mRNAs can effectively shift the axonal mRNA population. Consistent with this, the axonal RNA population shifts with development, with changes in growth state, and in response to extracellular stimulation. Each of these events must impact the transcriptional and transport apparatuses of the neuron, thus directly and indirectly modifying the axonal transcriptome.
Collapse
Affiliation(s)
- Cynthia Gomes
- Department of Biology, Drexel University, Philadelphia, Pennsylvania 19104 USA
| | - Tanuja T. Merianda
- Department of Biology, Drexel University, Philadelphia, Pennsylvania 19104 USA
| | - Seung Joon Lee
- Department of Biology, Drexel University, Philadelphia, Pennsylvania 19104 USA
| | - Soonmoon Yoo
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803 USA
| | - Jeffery L. Twiss
- Department of Biology, Drexel University, Philadelphia, Pennsylvania 19104 USA
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29201
| |
Collapse
|
58
|
Evolutionary conservation and expression of human RNA-binding proteins and their role in human genetic disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 825:1-55. [PMID: 25201102 DOI: 10.1007/978-1-4939-1221-6_1] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RNA-binding proteins (RBPs) are effectors and regulators of posttranscriptional gene regulation (PTGR). RBPs regulate stability, maturation, and turnover of all RNAs, often binding thousands of targets at many sites. The importance of RBPs is underscored by their dysregulation or mutations causing a variety of developmental and neurological diseases. This chapter globally discusses human RBPs and provides a brief introduction to their identification and RNA targets. We review RBPs based on common structural RNA-binding domains, study their evolutionary conservation and expression, and summarize disease associations of different RBP classes.
Collapse
|
59
|
Goodwin M, Swanson MS. RNA-binding protein misregulation in microsatellite expansion disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 825:353-88. [PMID: 25201111 PMCID: PMC4483269 DOI: 10.1007/978-1-4939-1221-6_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RNA-binding proteins (RBPs) play pivotal roles in multiple cellular pathways from transcription to RNA turnover by interacting with RNA sequence and/or structural elements to form distinct RNA-protein complexes. Since these complexes are required for the normal regulation of gene expression, mutations that alter RBP functions may result in a cascade of deleterious events that lead to severe disease. Here, we focus on a group of hereditary disorders, the microsatellite expansion diseases, which alter RBP activities and result in abnormal neurological and neuromuscular phenotypes. While many of these diseases are classified as adult-onset disorders, mounting evidence indicates that disruption of normal RNA-protein interaction networks during embryogenesis modifies developmental pathways, which ultimately leads to disease manifestations later in life. Efforts to understand the molecular basis of these disorders has already uncovered novel pathogenic mechanisms, including RNA toxicity and repeat-associated non-ATG (RAN) translation, and current studies suggest that additional surprising insights into cellular regulatory pathways will emerge in the future.
Collapse
Affiliation(s)
- Marianne Goodwin
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Cancer Genetics Research Complex, 2033 Mowry Road, Gainesville, FL, 32610-3610, USA
| | | |
Collapse
|
60
|
Jahromi AH, Fu Y, Miller KA, Nguyen L, Luu LM, Baranger AM, Zimmerman SC. Developing bivalent ligands to target CUG triplet repeats, the causative agent of myotonic dystrophy type 1. J Med Chem 2013; 56:9471-9481. [PMID: 24188018 DOI: 10.1021/jm400794z] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An expanded CUG repeat transcript (CUG(exp)) is the causative agent of myotonic dystrophy type 1 (DM1) by sequestering muscleblind-like 1 protein (MBNL1), a regulator of alternative splicing. On the basis of a ligand (1) that was previously reported to be active in an in vitro assay, we present the synthesis of a small library containing 10 dimeric ligands (4-13) that differ in length, composition, and attachment point of the linking chain. The oligoamino linkers gave a greater gain in affinity for CUG RNA and were more effective when compared to oligoether linkers. The most potent in vitro ligand (9) was shown to be aqueous-soluble and both cell- and nucleus-permeable, displaying almost complete dispersion of MBNL1 ribonuclear foci in a DM1 cell model. Direct evidence for the bioactivity of 9 was observed in its ability to disperse ribonuclear foci in individual live DM1 model cells using time-lapse confocal fluorescence microscopy.
Collapse
Affiliation(s)
- Amin Haghighat Jahromi
- Center for Biophysics and Computational Biology, University of Illinois, Urbana, IL, USA.,Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Yuan Fu
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Kali A Miller
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Lien Nguyen
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Long M Luu
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Anne M Baranger
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Steven C Zimmerman
- Center for Biophysics and Computational Biology, University of Illinois, Urbana, IL, USA.,Department of Chemistry, University of Illinois, Urbana, IL, USA
| |
Collapse
|
61
|
Donnelly CJ, Zhang PW, Pham JT, Heusler AR, Mistry NA, Vidensky S, Daley EL, Poth EM, Hoover B, Fines DM, Maragakis N, Tienari PJ, Petrucelli L, Traynor BJ, Wang J, Rigo F, Bennett CF, Blackshaw S, Sattler R, Rothstein JD. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 2013; 80:415-28. [PMID: 24139042 PMCID: PMC4098943 DOI: 10.1016/j.neuron.2013.10.015] [Citation(s) in RCA: 701] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2013] [Indexed: 12/11/2022]
Abstract
A hexanucleotide GGGGCC repeat expansion in the noncoding region of the C9ORF72 gene is the most common genetic abnormality in familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The function of the C9ORF72 protein is unknown, as is the mechanism by which the repeat expansion could cause disease. Induced pluripotent stem cell (iPSC)-differentiated neurons from C9ORF72 ALS patients revealed disease-specific (1) intranuclear GGGGCCexp RNA foci, (2) dysregulated gene expression, (3) sequestration of GGGGCCexp RNA binding protein ADARB2, and (4) susceptibility to excitotoxicity. These pathological and pathogenic characteristics were confirmed in ALS brain and were mitigated with antisense oligonucleotide (ASO) therapeutics to the C9ORF72 transcript or repeat expansion despite the presence of repeat-associated non-ATG translation (RAN) products. These data indicate a toxic RNA gain-of-function mechanism as a cause of C9ORF72 ALS and provide candidate antisense therapeutics and candidate human pharmacodynamic markers for therapy.
Collapse
Affiliation(s)
- Christopher J. Donnelly
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Ping-Wu Zhang
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Jacqueline T. Pham
- Department of Cellular and Molecular Medicine, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Aaron R. Heusler
- Department of Biochemistry and Molecular Biology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Nipun A. Mistry
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Svetlana Vidensky
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Elizabeth L. Daley
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Erin M. Poth
- Department of Neuroscience, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Benjamin Hoover
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Daniel M. Fines
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Nicholas Maragakis
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Pentti J. Tienari
- Biomedicum, Research Programs Unit, Molecular Neurology, University of Helsinki; Helsinki University Central Hospital, Department of Neurology, Haartmaninkatu 8, FIN-00290 Helsinki, Finland
| | - Leonard Petrucelli
- Department of Molecular Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Bryan J. Traynor
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institute of Health, 35 Convent Drive, Room 1A-1000, Bethesda, MD 20892, USA
| | - Jiou Wang
- Department of Neuroscience, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Frank Rigo
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - C. Frank Bennett
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Rita Sattler
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| | - Jeffrey D. Rothstein
- Department of Neurology, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Department of Cellular and Molecular Medicine, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
- Brain Science Institute, Johns Hopkins University, 855 N Wolfe Street, Rangos 2–270, Baltimore, MD 21205, USA
| |
Collapse
|
62
|
Hagerman R, Hagerman P. Advances in clinical and molecular understanding of the FMR1 premutation and fragile X-associated tremor/ataxia syndrome. Lancet Neurol 2013; 12:786-98. [PMID: 23867198 DOI: 10.1016/s1474-4422(13)70125-x] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fragile X syndrome, the most common heritable form of cognitive impairment, is caused by epigenetic silencing of the fragile X (FMR1) gene owing to large expansions (>200 repeats) of a non-coding CGG-repeat element. Smaller, so-called premutation expansions (55-200 repeats) can cause a family of neurodevelopmental phenotypes (attention deficit hyperactivity disorder, autism spectrum disorder, seizure disorder) and neurodegenerative (fragile X-associated tremor/ataxia syndrome [FXTAS]) phenotypes through an entirely distinct molecular mechanism involving increased FMR1 mRNA production and toxicity. Results of basic cellular, animal, and human studies have helped to elucidate the underlying RNA toxicity mechanism, while clinical research is providing a more nuanced picture of the range of clinical manifestations. Advances of knowledge on both mechanistic and clinical fronts are driving new approaches to targeted treatment, but two important necessities are emerging: to define the extent to which the mechanisms contributing to FXTAS also contribute to other neurodegenerative and medical disorders, and to redefine FXTAS in view of its differing presentations and associated features.
Collapse
Affiliation(s)
- Randi Hagerman
- Department of Pediatrics and the MIND Institute, University of California, Davis, School of Medicine, Davis, CA, USA
| | | |
Collapse
|
63
|
Abstract
Well-established rules of translational initiation have been used as a cornerstone in molecular biology to understand gene expression and to frame fundamental questions on what proteins a cell synthesizes, how proteins work and to predict the consequences of mutations. For a group of neurological diseases caused by the abnormal expansion of short segments of DNA (e.g. CAG•CTG repeats), mutations within or outside of predicted coding and non-coding regions are thought to cause disease by protein gain- or loss-of-function or RNA gain-of-function mechanisms. In contrast to these predictions, the recent discovery of repeat-associated non-ATG (RAN) translation showed expansion mutations can express homopolymeric expansion proteins in all three reading frames without an AUG start codon. This unanticipated, non-canonical type of protein translation is length-and hairpin-dependent, takes place without frameshifting or RNA editing and occurs across a variety of repeat motifs. To date, RAN proteins have been reported in spinocerebellar ataxia type 8 (SCA8), myotonic dystrophy type 1 (DM1), fragile X tremor ataxia syndrome (FXTAS) and C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). In this article, we review what is currently known about RAN translation and recent progress toward understanding its contribution to disease.
Collapse
Affiliation(s)
| | - Laura P.W. Ranum
- To whom correspondence should be addressed at: Director, Center for NeuroGenetics, Professor of Molecular Genetics and Microbiology, College of Medicine, University of Florida, 2033 Mowry Road, PO Box 103610, Gainesville, FL 32610-3610, USA. Tel: +1-3522945209; Fax: +1-3522738284;
| |
Collapse
|
64
|
Current insights into the C9orf72 repeat expansion diseases of the FTLD/ALS spectrum. Trends Neurosci 2013; 36:450-9. [DOI: 10.1016/j.tins.2013.04.010] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 04/28/2013] [Accepted: 04/30/2013] [Indexed: 11/20/2022]
|
65
|
Feng D, Xie J. Aberrant splicing in neurological diseases. WILEY INTERDISCIPLINARY REVIEWS-RNA 2013; 4:631-49. [PMID: 23821330 DOI: 10.1002/wrna.1184] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 12/12/2022]
Abstract
Splicing of precursor messenger RNA (pre-mRNA) removes the intervening sequences (introns) and joins the expressed regions (exons) in the nucleus, before an intron-containing eukaryotic mRNA transcript can be exported and translated into proteins in the cytoplasm. While some sequences are always included or excluded (constitutive splicing), others can be selectively used (alternative splicing) in this process. Particularly by alternative splicing, up to tens of thousands of variant transcripts can be produced from a single gene, which contributes greatly to the proteomic diversity for such complex cellular functions as 'wiring' neurons in the nervous system. Disruption of this process leads to aberrant splicing, which accounts for the defects of up to 50% of mutations that cause certain human genetic diseases. In this review, we describe the different mechanisms of aberrant splicing that cause or have been associated with neurological diseases.
Collapse
Affiliation(s)
- Dairong Feng
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
66
|
Fragile X-associated tremor/ataxia syndrome (FXTAS): pathology and mechanisms. Acta Neuropathol 2013; 126:1-19. [PMID: 23793382 DOI: 10.1007/s00401-013-1138-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/30/2013] [Indexed: 12/17/2022]
Abstract
Since its discovery in 2001, our understanding of fragile X-associated tremor/ataxia syndrome (FXTAS) has undergone a remarkable transformation. Initially characterized rather narrowly as an adult-onset movement disorder, the definition of FXTAS is broadening; moreover, the disorder is now recognized as only one facet of a much broader clinical pleiotropy among children and adults who carry premutation alleles of the FMR1 gene. Furthermore, the intranuclear inclusions of FXTAS, once thought to be a CNS-specific marker of the disorder, are now known to be widely distributed in multiple non-CNS tissues; this observation fundamentally changes our concept of the disease, and may provide the basis for understanding the diverse medical problems associated with the premutation. Recent work on the pathogenic mechanisms underlying FXTAS indicates that the origins of the late-onset neurodegenerative disorder actually lie in early development, raising the likelihood that all forms of clinical involvement among premutation carriers have a common underlying mechanistic basis. There has also been great progress in our understanding of the triggering event(s) in FXTAS pathogenesis, which is now thought to involve sequestration of one or more nuclear proteins involved with microRNA biogenesis. Moreover, there is mounting evidence that mitochondrial dysregulation contributes to the decreased cell function and loss of viability, evident in mice even during the neonatal period. Taken together, these recent findings offer hope for early interventions for FXTAS, well before the onset of overt disease, and for the treatment of other forms of clinical involvement among premutation carriers.
Collapse
|
67
|
Abstract
The process of misfolding of proteins that can trigger a pathogenic cascade leading to neurodegenerative diseases largely originates intracellularly. It is possible to harness the specificity and affinity of antibodies to counteract either protein misfolding itself, or the aberrant interactions and excess stressors immediately downstream of the primary insult. This review covers the emerging field of engineering intracellular antibody fragments, intrabodies and nanobodies, in neurodegeneration. Huntington's disease has provided the clearest proof of concept for this approach. The model systems and readouts for this disorder power the studies, and the potential to intervene therapeutically at early stages in known carriers with projected ages of onset increases the chances of meaningful clinical trials. Both single-chain Fv and single-domain nanobodies have been identified against specific targets; data have allowed feedback for rational design of bifunctional constructs, as well as target validation. Intrabodies that can modulate the primary accumulating protein in Parkinson's disease, alpha-synuclein, are also reviewed, covering a range of domains and conformers. Recombinant antibody technology has become a major player in the therapeutic pipeline for cancer, infectious diseases, and autoimmunity. There is also tremendous potential for applying this powerful biotechnology to neurological diseases.
Collapse
Affiliation(s)
- Anne Messer
- New York State Dept of Health, Wadsworth Center, Albany, NY 12208, USA.
| | | |
Collapse
|
68
|
Fiszer A, Krzyzosiak WJ. RNA toxicity in polyglutamine disorders: concepts, models, and progress of research. J Mol Med (Berl) 2013; 91:683-91. [PMID: 23512265 PMCID: PMC3659269 DOI: 10.1007/s00109-013-1016-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 02/19/2013] [Accepted: 02/25/2013] [Indexed: 01/13/2023]
Abstract
In Huntington's disease and other polyglutamine (polyQ) disorders, mutant proteins containing a long polyQ stretch are well documented as the trigger of numerous aberrant cellular processes that primarily lead to degeneration and, ultimately, the death of neuronal cells. However, mutant transcripts containing expanded CAG repeats may also be toxic and contribute to cellular dysfunction. The exact nature and importance of RNA toxicity in polyQ diseases are only beginning to be recognized, and the first insights have mainly resulted from studies using simple model systems. In this review, we briefly present the basic mechanisms of protein toxicity in polyQ disorders and RNA toxicity in myotonic dystrophy type 1 and discuss recent results suggesting that the pathogenesis of polyQ diseases may also be mediated by mutant transcripts. This review is focused on the experimental systems used thus far to demonstrate RNA toxicity in polyQ disorders and the design of new systems that will be more relevant to the human disease situation and capable of separating RNA toxicity from protein toxicity.
Collapse
Affiliation(s)
- Agnieszka Fiszer
- Laboratory of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Wlodzimierz J. Krzyzosiak
- Laboratory of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
69
|
Jahromi AH, Nguyen L, Fu Y, Miller KA, Baranger AM, Zimmerman SC. A novel CUG(exp)·MBNL1 inhibitor with therapeutic potential for myotonic dystrophy type 1. ACS Chem Biol 2013; 8:1037-43. [PMID: 23480597 DOI: 10.1021/cb400046u] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is caused by an expanded CUG repeat (CUG(exp)) that sequesters muscleblind-like 1 protein (MBNL1), a protein that regulates alternative splicing. CUG(exp) RNA is a validated drug target for this currently untreatable disease. Herein, we develop a bioactive small molecule (1) that targets CUG(exp) RNA and is able to inhibit the CUG(exp)·MBNL1 interaction in cells that model DM1. The core of this small molecule is based on ligand 2, which was previously reported to be active in an in vitro assay. A polyamine-derivative side chain was conjugated to this core to make it aqueous-soluble and cell-penetrable. In a DM1 cell model this conjugate was found to disperse CUG(exp) ribonuclear foci, release MBNL1, and partially reverse the mis-splicing of the insulin receptor pre-mRNA. Direct evidence for ribonuclear foci dispersion by this ligand was obtained in a live DM1 cell model using time-lapse confocal microscopy.
Collapse
Affiliation(s)
- Amin Haghighat Jahromi
- Center for Biophysics
and Computational Biology and ‡Department of Chemistry, University of Illinois, Urbana, Illinois, United States
| | - Lien Nguyen
- Center for Biophysics
and Computational Biology and ‡Department of Chemistry, University of Illinois, Urbana, Illinois, United States
| | - Yuan Fu
- Center for Biophysics
and Computational Biology and ‡Department of Chemistry, University of Illinois, Urbana, Illinois, United States
| | - Kali A. Miller
- Center for Biophysics
and Computational Biology and ‡Department of Chemistry, University of Illinois, Urbana, Illinois, United States
| | - Anne M. Baranger
- Center for Biophysics
and Computational Biology and ‡Department of Chemistry, University of Illinois, Urbana, Illinois, United States
| | - Steven C. Zimmerman
- Center for Biophysics
and Computational Biology and ‡Department of Chemistry, University of Illinois, Urbana, Illinois, United States
| |
Collapse
|
70
|
Haghighat Jahromi A, Honda M, Zimmerman SC, Spies M. Single-molecule study of the CUG repeat-MBNL1 interaction and its inhibition by small molecules. Nucleic Acids Res 2013; 41:6687-97. [PMID: 23661680 PMCID: PMC3711446 DOI: 10.1093/nar/gkt330] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Effective drug discovery and optimization can be accelerated by techniques capable of deconvoluting the complexities often present in targeted biological systems. We report a single-molecule approach to study the binding of an alternative splicing regulator, muscleblind-like 1 protein (MBNL1), to (CUG)n = 4,6 and the effect of small molecules on this interaction. Expanded CUG repeats (CUG(exp)) are the causative agent of myotonic dystrophy type 1 by sequestering MBNL1. MBNL1 is able to bind to the (CUG)n-inhibitor complex, indicating that the inhibition is not a straightforward competitive process. A simple ligand, highly selective for CUG(exp), was used to design a new dimeric ligand that binds to (CUG)n almost 50-fold more tightly and is more effective in destabilizing MBNL1-(CUG)4. The single-molecule method and the analysis framework might be extended to the study of other biomolecular interactions.
Collapse
Affiliation(s)
- Amin Haghighat Jahromi
- Center for Biophysics and Computational Biology, University of Illinois, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
71
|
Nelson DL, Orr HT, Warren ST. The unstable repeats--three evolving faces of neurological disease. Neuron 2013; 77:825-43. [PMID: 23473314 PMCID: PMC3608403 DOI: 10.1016/j.neuron.2013.02.022] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2013] [Indexed: 01/08/2023]
Abstract
Disorders characterized by expansion of an unstable nucleotide repeat account for a number of inherited neurological diseases. Here, we review examples of unstable repeat disorders that nicely illustrate three of the major pathogenic mechanisms associated with these diseases: loss of function typically by disrupting transcription of the mutated gene, RNA toxic gain of function, and protein toxic gain of function. In addition to providing insight into the mechanisms underlying these devastating neurological disorders, the study of these unstable microsatellite repeat disorders has provided insight into very basic aspects of neuroscience.
Collapse
Affiliation(s)
- David L. Nelson
- Department of Molecular and Human Genetics, Baylor College
of Medicine, Houston, TX 77030
| | - Harry T. Orr
- Department of Laboratory Medicine and Pathology, University
of Minnesota, Minneapolis, MN 55455
| | - Stephen T. Warren
- Department of Human Genetics, Emory University School of
Medicine, Atlanta, GA 30322
| |
Collapse
|
72
|
Abstract
Instability of repetitive DNA sequences within the genome is associated with a number of human diseases. The expansion of trinucleotide repeats is recognized as a major cause of neurological and neuromuscular diseases, and progress in understanding the mutations over the last 20 years has been substantial. Here we provide a brief summary of progress with an emphasis on technical advances at different stages.
Collapse
Affiliation(s)
- Helen Budworth
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | |
Collapse
|
73
|
Belzil VV, Gendron TF, Petrucelli L. RNA-mediated toxicity in neurodegenerative disease. Mol Cell Neurosci 2012; 56:406-19. [PMID: 23280309 DOI: 10.1016/j.mcn.2012.12.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/19/2012] [Accepted: 12/21/2012] [Indexed: 12/12/2022] Open
Abstract
Cellular viability depends upon the well-orchestrated functions carried out by numerous protein-coding and non-coding RNAs, as well as RNA-binding proteins. During the last decade, it has become increasingly evident that abnormalities in RNA processing represent a common feature among many neurodegenerative diseases. In "RNAopathies", which include diseases caused by non-coding repeat expansions, RNAs exert toxicity via diverse mechanisms: RNA foci formation, bidirectional transcription, and the production of toxic RNAs and proteins by repeat associated non-ATG translation. The mechanisms of toxicity in "RNA-binding proteinopathies", diseases in which RNA-binding proteins like TDP-43 and FUS play a prominent role, have yet to be fully elucidated. Nonetheless, both loss of function of the RNA binding protein, and a toxic gain of function resulting from its aggregation, are thought to be involved in disease pathogenesis. As part of the special issue on RNA and Splicing Regulation in Neurodegeneration, this review intends to explore the diverse RNA-related mechanisms contributing to neurodegeneration, with a special emphasis on findings emerging from animal models.
Collapse
Affiliation(s)
- Veronique V Belzil
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | | |
Collapse
|