Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 22(9): 589-607, 2021 09.
Article in English | MEDLINE | ID: mdl-34140671

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.


Subject(s)
DNA Repeat Expansion/genetics , Neurodegenerative Diseases/genetics , 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
2.
Mol Cell ; 83(3): 324-329, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36736306

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.


Subject(s)
Amyotrophic Lateral Sclerosis , Huntington Disease , Myotonic Dystrophy , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Proteins/genetics , Huntington Disease/genetics , RNA/genetics , Myotonic Dystrophy/genetics , Trinucleotide Repeat Expansion/genetics
5.
Cell ; 150(4): 710-24, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22901804

ABSTRACT

The muscleblind-like (Mbnl) family of RNA-binding proteins plays important roles in muscle and eye development and in myotonic dystrophy (DM), in which expanded CUG or CCUG repeats functionally deplete Mbnl proteins. We identified transcriptome-wide functional and biophysical targets of Mbnl proteins in brain, heart, muscle, and myoblasts by using RNA-seq and CLIP-seq approaches. This analysis identified several hundred splicing events whose regulation depended on Mbnl function in a pattern indicating functional interchangeability between Mbnl1 and Mbnl2. A nucleotide resolution RNA map associated repression or activation of exon splicing with Mbnl binding near either 3' splice site or near the downstream 5' splice site, respectively. Transcriptomic analysis of subcellular compartments uncovered a global role for Mbnls in regulating localization of mRNAs in both mouse and Drosophila cells, and Mbnl-dependent translation and protein secretion were observed for a subset of mRNAs with Mbnl-dependent localization. These findings hold several new implications for DM pathogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Myotonic Dystrophy/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Transcriptome , 3' Untranslated Regions , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins , Drosophila melanogaster/metabolism , Exons , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Myoblasts/metabolism , Myotonic Dystrophy/genetics , Nuclear Proteins , Organ Specificity , RNA Splice Sites , RNA-Binding Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 121(19): e2321438121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38687782

ABSTRACT

Successful CRISPR/Cas9-based gene editing in skeletal muscle is dependent on efficient propagation of Cas9 to all myonuclei in the myofiber. However, nuclear-targeted gene therapy cargos are strongly restricted to their myonuclear domain of origin. By screening nuclear localization signals and nuclear export signals, we identify "Myospreader," a combination of short peptide sequences that promotes myonuclear propagation. Appending Myospreader to Cas9 enhances protein stability and myonuclear propagation in myoblasts and myofibers. AAV-delivered Myospreader dCas9 better inhibits transcription of toxic RNA in a myotonic dystrophy mouse model. Furthermore, Myospreader Cas9 achieves higher rates of gene editing in CRISPR reporter and Duchenne muscular dystrophy mouse models. Myospreader reveals design principles relevant to all nuclear-targeted gene therapies and highlights the importance of the spatial dimension in therapeutic development.


Subject(s)
CRISPR-Cas Systems , Cell Nucleus , Gene Editing , Genetic Therapy , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Gene Editing/methods , Animals , Mice , Muscle, Skeletal/metabolism , Cell Nucleus/metabolism , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/genetics , Humans , Nuclear Localization Signals/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , Disease Models, Animal , Myoblasts/metabolism
7.
Genes Dev ; 33(23-24): 1635-1640, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31624084

ABSTRACT

Short tandem repeats (STRs) are prone to expansion mutations that cause multiple hereditary neurological and neuromuscular diseases. To study pathomechanisms using mouse models that recapitulate the tissue specificity and developmental timing of an STR expansion gene, we used rolling circle amplification and CRISPR/Cas9-mediated genome editing to generate Dmpk CTG expansion (CTGexp) knockin models of myotonic dystrophy type 1 (DM1). We demonstrate that skeletal muscle myoblasts and brain choroid plexus epithelial cells are particularly susceptible to Dmpk CTGexp mutations and RNA missplicing. Our results implicate dysregulation of muscle regeneration and cerebrospinal fluid homeostasis as early pathogenic events in DM1.


Subject(s)
Alternative Splicing/genetics , Microsatellite Repeats/genetics , Muscle, Skeletal/physiopathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/physiopathology , RNA Splicing/genetics , 3' Untranslated Regions/genetics , Animals , Choroid Plexus/physiopathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/cytology , Mutation , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , RNA-Binding Proteins/genetics
8.
Mol Cell ; 68(3): 479-490.e5, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29056323

ABSTRACT

Transcription of expanded microsatellite repeats is associated with multiple human diseases, including myotonic dystrophy, Fuchs endothelial corneal dystrophy, and C9orf72-ALS/FTD. Reducing production of RNA and proteins arising from these expanded loci holds therapeutic benefit. Here, we tested the hypothesis that deactivated Cas9 enzyme impedes transcription across expanded microsatellites. We observed a repeat length-, PAM-, and strand-dependent reduction of repeat-containing RNAs upon targeting dCas9 directly to repeat sequences; targeting the non-template strand was more effective. Aberrant splicing patterns were rescued in DM1 cells, and production of RAN peptides characteristic of DM1, DM2, and C9orf72-ALS/FTD cells was drastically decreased. Systemic delivery of dCas9/gRNA by adeno-associated virus led to reductions in pathological RNA foci, rescue of chloride channel 1 protein expression, and decreased myotonia. These observations suggest that transcription of microsatellite repeat-containing RNAs is more sensitive to perturbation than transcription of other RNAs, indicating potentially viable strategies for therapeutic intervention.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Endonucleases/metabolism , Genetic Therapy/methods , Microsatellite Repeats , Myotonic Dystrophy/therapy , Transcription, Genetic , Alternative Splicing , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Dependovirus/genetics , Disease Models, Animal , Down-Regulation , Enzyme Activation , Female , Genetic Vectors , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Transgenic , Myoblasts/metabolism , Myoblasts/pathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA, Guide, Kinetoplastida/biosynthesis , RNA, Guide, Kinetoplastida/genetics , Transduction, Genetic , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
9.
Genes Dev ; 31(11): 1122-1133, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28698297

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a CTG microsatellite expansion (CTGexp) disorder caused by expression of CUGexp RNAs. These mutant RNAs alter the activities of RNA processing factors, including MBNL proteins, leading to re-expression of fetal isoforms in adult tissues and DM1 pathology. While this pathogenesis model accounts for adult-onset disease, the molecular basis of congenital DM (CDM) is unknown. Here, we test the hypothesis that disruption of developmentally regulated RNA alternative processing pathways contributes to CDM disease. We identify prominent alternative splicing and polyadenylation abnormalities in infant CDM muscle, and, although most are also misregulated in adult-onset DM1, dysregulation is significantly more severe in CDM. Furthermore, analysis of alternative splicing during human myogenesis reveals that CDM-relevant exons undergo prenatal RNA isoform transitions and are predicted to be disrupted by CUGexp-associated mechanisms in utero. To test this possibility and the contribution of MBNLs to CDM pathogenesis, we generated mouse Mbnl double (Mbnl1; Mbnl2) and triple (Mbnl1; Mbnl2; Mbnl3) muscle-specific knockout models that recapitulate the congenital myopathy, gene expression, and spliceopathy defects characteristic of CDM. This study demonstrates that RNA misprocessing is a major pathogenic factor in CDM and provides novel mouse models to further examine roles for cotranscriptional/post-transcriptional gene regulation during development.


Subject(s)
Muscle Development/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/physiopathology , RNA Processing, Post-Transcriptional/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Animals , Carrier Proteins/genetics , Cells, Cultured , Child, Preschool , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Gene Knockout Techniques , Humans , Infant , Mice , RNA-Binding Proteins/metabolism
10.
Hum Mol Genet ; 31(18): 3144-3160, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35567413

ABSTRACT

Myotonic dystrophy (DM) is caused by expansions of C(C)TG repeats in the non-coding regions of the DMPK and CNBP genes, and DM patients often suffer from sudden cardiac death due to lethal conduction block or arrhythmia. Specific molecular changes that underlie DM cardiac pathology have been linked to repeat-associated depletion of Muscleblind-like (MBNL) 1 and 2 proteins and upregulation of CUGBP, Elav-like family member 1 (CELF1). Hypothesis solely targeting MBNL1 or CELF1 pathways that could address all the consequences of repeat expansion in heart remained inconclusive, particularly when the direct cause of mortality and results of transcriptome analyses remained undetermined in Mbnl compound knockout (KO) mice with cardiac phenotypes. Here, we develop Myh6-Cre double KO (DKO) (Mbnl1-/-; Mbnl2cond/cond; Myh6-Cre+/-) mice to eliminate Mbnl1/2 in cardiomyocytes and observe spontaneous lethal cardiac events under no anesthesia. RNA sequencing recapitulates DM heart spliceopathy and shows gene expression changes that were previously undescribed in DM heart studies. Notably, immunoblotting reveals a nearly 6-fold increase of Calsequestrin 1 and 50% reduction of epidermal growth factor proteins. Our findings demonstrate that complete ablation of MBNL1/2 in cardiomyocytes is essential for generating sudden death due to lethal cardiac rhythms and reveal potential mechanisms for DM heart pathogenesis.


Subject(s)
Myotonic Dystrophy , Alternative Splicing/genetics , Animals , Calsequestrin/genetics , DNA-Binding Proteins/genetics , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , EGF Family of Proteins/genetics , EGF Family of Proteins/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Myotonic Dystrophy/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
11.
Brain ; 146(10): 4217-4232, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37143315

ABSTRACT

Myotonic dystrophy type 1 is a dominantly inherited multisystemic disease caused by CTG tandem repeat expansions in the DMPK 3' untranslated region. These expanded repeats are transcribed and produce toxic CUG RNAs that sequester and inhibit activities of the MBNL family of developmental RNA processing factors. Although myotonic dystrophy is classified as a muscular dystrophy, the brain is also severely affected by an unusual cohort of symptoms, including hypersomnia, executive dysfunction, as well as early onsets of tau/MAPT pathology and cerebral atrophy. To address the molecular and cellular events that lead to these pathological outcomes, we recently generated a mouse Dmpk CTG expansion knock-in model and identified choroid plexus epithelial cells as particularly affected by the expression of toxic CUG expansion RNAs. To determine if toxic CUG RNAs perturb choroid plexus functions, alternative splicing analysis was performed on lateral and hindbrain choroid plexi from Dmpk CTG knock-in mice. Choroid plexus transcriptome-wide changes were evaluated in Mbnl2 knockout mice, a developmental-onset model of myotonic dystrophy brain dysfunction. To determine if transcriptome changes also occurred in the human disease, we obtained post-mortem choroid plexus for RNA-seq from neurologically unaffected (two females, three males; ages 50-70 years) and myotonic dystrophy type 1 (one female, three males; ages 50-70 years) donors. To test that choroid plexus transcriptome alterations resulted in altered CSF composition, we obtained CSF via lumbar puncture from patients with myotonic dystrophy type 1 (five females, five males; ages 35-55 years) and non-myotonic dystrophy patients (three females, four males; ages 26-51 years), and western blot and osmolarity analyses were used to test CSF alterations predicted by choroid plexus transcriptome analysis. We determined that CUG RNA induced toxicity was more robust in the lateral choroid plexus of Dmpk CTG knock-in mice due to comparatively higher Dmpk and lower Mbnl RNA levels. Impaired transitions to adult splicing patterns during choroid plexus development were identified in Mbnl2 knockout mice, including mis-splicing previously found in Dmpk CTG knock-in mice. Whole transcriptome analysis of myotonic dystrophy type 1 choroid plexus revealed disease-associated RNA expression and mis-splicing events. Based on these RNA changes, predicted alterations in ion homeostasis, secretory output and CSF composition were confirmed by analysis of myotonic dystrophy type 1 CSF. Our results implicate choroid plexus spliceopathy and concomitant alterations in CSF homeostasis as an unappreciated contributor to myotonic dystrophy type 1 CNS pathogenesis.


Subject(s)
Myotonic Dystrophy , Humans , Female , Mice , Animals , Myotonic Dystrophy/genetics , Choroid Plexus/metabolism , Choroid Plexus/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Alternative Splicing , RNA/genetics , Mice, Knockout , Trinucleotide Repeat Expansion
12.
Mol Cell ; 61(6): 821-33, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26907613

ABSTRACT

Spatial restriction of mRNA to distinct subcellular locations enables local regulation and synthesis of proteins. However, the organizing principles of mRNA localization remain poorly understood. Here we analyzed subcellular transcriptomes of neural projections and soma of primary mouse cortical neurons and two neuronal cell lines and found that alternative last exons (ALEs) often confer isoform-specific localization. Surprisingly, gene-distal ALE isoforms were four times more often localized to neurites than gene-proximal isoforms. Localized isoforms were induced during neuronal differentiation and enriched for motifs associated with muscleblind-like (Mbnl) family RNA-binding proteins. Depletion of Mbnl1 and/or Mbnl2 reduced localization of hundreds of transcripts, implicating Mbnls in localization of mRNAs to neurites. We provide evidence supporting a model in which the linkage between genomic position of ALEs and subcellular localization enables coordinated induction of localization-competent mRNA isoforms through a post-transcriptional regulatory program that is induced during differentiation and reversed in cellular reprogramming and cancer.


Subject(s)
DNA-Binding Proteins/genetics , Neurites/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Alternative Splicing/genetics , Animals , Cell Differentiation/genetics , Cellular Reprogramming/genetics , DNA-Binding Proteins/antagonists & inhibitors , Exons , Gene Expression Regulation, Developmental , Humans , Mice , Protein Isoforms , Protein Structure, Tertiary , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/antagonists & inhibitors , Transcriptome/genetics
13.
J Physiol ; 601(4): 723-741, 2023 02.
Article in English | MEDLINE | ID: mdl-36629254

ABSTRACT

Most cells in the body are mononuclear whereas skeletal muscle fibres are uniquely multinuclear. The nuclei of muscle fibres (myonuclei) are usually situated peripherally which complicates the equitable distribution of gene products. Myonuclear abundance can also change under conditions such as hypertrophy and atrophy. Specialised zones in muscle fibres have different functions and thus distinct synthetic demands from myonuclei. The complex structure and regulatory requirements of multinuclear muscle cells understandably led to the hypothesis that myonuclei govern defined 'domains' to maintain homeostasis and facilitate adaptation. The purpose of this review is to provide historical context for the myonuclear domain and evaluate its veracity with respect to mRNA and protein distribution resulting from myonuclear transcription. We synthesise insights from past and current in vitro and in vivo genetically modified models for studying the myonuclear domain under dynamic conditions. We also cover the most contemporary knowledge on mRNA and protein transport in muscle cells. Insights from emerging technologies such as single myonuclear RNA-sequencing further inform our discussion of the myonuclear domain. We broadly conclude: (1) the myonuclear domain can be flexible during muscle fibre growth and atrophy, (2) the mechanisms and role of myonuclear loss and motility deserve further consideration, (3) mRNA in muscle is actively transported via microtubules and locally restricted, but proteins may travel far from a myonucleus of origin and (4) myonuclear transcriptional specialisation extends beyond the classic neuromuscular and myotendinous populations. A deeper understanding of the myonuclear domain in muscle may promote effective therapies for ageing and disease.


Subject(s)
Muscle Fibers, Skeletal , Muscle, Skeletal , Adult , Humans , Muscle, Skeletal/physiology , Muscle Fibers, Skeletal/physiology , Cell Nucleus/metabolism , RNA, Messenger/metabolism , Atrophy/metabolism , Atrophy/pathology
14.
Hum Mol Genet ; 29(24): 3900-3918, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33378537

ABSTRACT

C9orf72 ALS/FTD patients show remarkable clinical heterogeneity, but the complex biology of the repeat expansion mutation has limited our understanding of the disease. BAC transgenic mice were used to better understand the molecular mechanisms and repeat length effects of C9orf72 ALS/FTD. Genetic analyses of these mice demonstrate that the BAC transgene and not integration site effects cause ALS/FTD phenotypes. Transcriptomic changes in cell proliferation, inflammation and neuronal pathways are found late in disease and alternative splicing changes provide early molecular markers that worsen with disease progression. Isogenic sublines of mice with 800, 500 or 50 G4C2 repeats generated from the single-copy C9-500 line show longer repeats result in earlier onset, increased disease penetrance and increased levels of RNA foci and dipeptide RAN protein aggregates. These data demonstrate G4C2 repeat length is an important driver of disease and identify alternative splicing changes as early biomarkers of C9orf72 ALS/FTD.


Subject(s)
Alternative Splicing , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/metabolism , DNA Repeat Expansion , Disease Models, Animal , Frontotemporal Dementia/pathology , Penetrance , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/metabolism , Animals , C9orf72 Protein/genetics , Frontotemporal Dementia/etiology , Frontotemporal Dementia/metabolism , Humans , Mice , Mice, Transgenic , Mutation , Phenotype
15.
Proc Natl Acad Sci U S A ; 117(5): 2406-2411, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31964809

ABSTRACT

As the area of small molecules interacting with RNA advances, general routes to provide bioactive compounds are needed as ligands can bind RNA avidly to sites that will not affect function. Small-molecule targeted RNA degradation will thus provide a general route to affect RNA biology. A non-oligonucleotide-containing compound was designed from sequence to target the precursor to oncogenic microRNA-21 (pre-miR-21) for enzymatic destruction with selectivity that can exceed that for protein-targeted medicines. The compound specifically binds the target and contains a heterocycle that recruits and activates a ribonuclease to pre-miR-21 to substoichiometrically effect its cleavage and subsequently impede metastasis of breast cancer to lung in a mouse model. Transcriptomic and proteomic analyses demonstrate that the compound is potent and selective, specifically modulating oncogenic pathways. Thus, small molecules can be designed from sequence to have all of the functional repertoire of oligonucleotides, including inducing enzymatic degradation, and to selectively and potently modulate RNA function in vivo.


Subject(s)
Breast Neoplasms/drug therapy , MicroRNAs/metabolism , Ribonucleases/metabolism , Small Molecule Libraries/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Design , Female , Humans , Mice , MicroRNAs/chemistry , Molecular Structure , Neoplasm Metastasis , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Ribonucleases/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
16.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30206144

ABSTRACT

Spinocerebellar ataxia type 8 (SCA8) is caused by a bidirectionally transcribed CTG·CAG expansion that results in the in vivo accumulation of CUG RNA foci, an ATG-initiated polyGln and a polyAla protein expressed by repeat-associated non-ATG (RAN) translation. Although RAN proteins have been reported in a growing number of diseases, the mechanisms and role of RAN translation in disease are poorly understood. We report a novel toxic SCA8 polySer protein which accumulates in white matter (WM) regions as aggregates that increase with age and disease severity. WM regions with polySer aggregates show demyelination and axonal degeneration in SCA8 human and mouse brains. Additionally, knockdown of the eukaryotic translation initiation factor eIF3F in cells reduces steady-state levels of SCA8 polySer and other RAN proteins. Taken together, these data show polySer and WM abnormalities contribute to SCA8 and identify eIF3F as a novel modulator of RAN protein accumulation.


Subject(s)
Aging/metabolism , Eukaryotic Initiation Factor-3/metabolism , Nerve Tissue Proteins/metabolism , Spinocerebellar Degenerations/metabolism , White Matter/metabolism , Aging/genetics , Aging/pathology , Animals , Eukaryotic Initiation Factor-3/genetics , HeLa Cells , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/pathology , White Matter/pathology
17.
Nucleic Acids Res ; 48(11): 5859-5872, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32421779

ABSTRACT

Subcellular organization of RNAs and proteins is critical for cell function, but we still lack global maps and conceptual frameworks for how these molecules are localized in cells and tissues. Here, we introduce ATLAS-Seq, which generates transcriptomes and proteomes from detergent-free tissue lysates fractionated across a sucrose gradient. Proteomic analysis of fractions confirmed separation of subcellular compartments. Unexpectedly, RNAs tended to co-sediment with other RNAs in similar protein complexes, cellular compartments, or with similar biological functions. With the exception of those encoding secreted proteins, most RNAs sedimented differently than their encoded protein counterparts. To identify RNA binding proteins potentially driving these patterns, we correlated their sedimentation profiles to all RNAs, confirming known interactions and predicting new associations. Hundreds of alternative RNA isoforms exhibited distinct sedimentation patterns across the gradient, despite sharing most of their coding sequence. These observations suggest that transcriptomes can be organized into networks of co-segregating mRNAs encoding functionally related proteins and provide insights into the establishment and maintenance of subcellular organization.


Subject(s)
Cell Fractionation , Cellular Microenvironment , Intracellular Space/chemistry , RNA/analysis , RNA/metabolism , Sequence Analysis, RNA , Transcriptome , Animals , Cell Extracts/chemistry , Centrifugation, Density Gradient , Female , Liver/cytology , Liver/metabolism , Mass Spectrometry , Mice , RNA/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribosomes/chemistry , Sucrose
18.
Proc Natl Acad Sci U S A ; 116(42): 20991-21000, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31570586

ABSTRACT

A CTG repeat expansion in the DMPK gene is the causative mutation of myotonic dystrophy type 1 (DM1). Transcription of the expanded CTG repeat produces toxic gain-of-function CUG RNA, leading to disease symptoms. A screening platform that targets production or stability of the toxic CUG RNA in a selective manner has the potential to provide new biological and therapeutic insights. A DM1 HeLa cell model was generated that stably expresses a toxic r(CUG)480 and an analogous r(CUG)0 control from DMPK and was used to measure the ratio-metric level of r(CUG)480 versus r(CUG)0. This DM1 HeLa model recapitulates pathogenic hallmarks of DM1, including CUG ribonuclear foci and missplicing of pre-mRNA targets of the muscleblind (MBNL) alternative splicing factors. Repeat-selective screening using this cell line led to the unexpected identification of multiple microtubule inhibitors as hits that selectively reduce r(CUG)480 levels and partially rescue MBNL-dependent missplicing. These results were validated by using the Food and Drug Administration-approved clinical microtubule inhibitor colchicine in DM1 mouse and primary patient cell models. The mechanism of action was found to involve selective reduced transcription of the CTG expansion that we hypothesize to involve the LINC (linker of nucleoskeleton and cytoskeleton) complex. The unanticipated identification of microtubule inhibitors as selective modulators of toxic CUG RNA opens research directions for this form of muscular dystrophy and may shed light on the biology of CTG repeat expansion and inform therapeutic avenues. This approach has the potential to identify modulators of expanded repeat-containing gene expression for over 30 microsatellite expansion disorders.


Subject(s)
Drug Evaluation, Preclinical/methods , Microtubules/drug effects , Myotonic Dystrophy/genetics , RNA/genetics , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Animals , HeLa Cells , Humans , Mice , Mice, Transgenic , Microtubules/genetics , Microtubules/metabolism , Myotonic Dystrophy/enzymology , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , RNA/chemistry , RNA/metabolism
19.
Proc Natl Acad Sci U S A ; 116(16): 7799-7804, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926669

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an incurable neuromuscular disorder caused by an expanded CTG repeat that is transcribed into r(CUG)exp The RNA repeat expansion sequesters regulatory proteins such as Muscleblind-like protein 1 (MBNL1), which causes pre-mRNA splicing defects. The disease-causing r(CUG)exp has been targeted by antisense oligonucleotides, CRISPR-based approaches, and RNA-targeting small molecules. Herein, we describe a designer small molecule, Cugamycin, that recognizes the structure of r(CUG)exp and cleaves it in both DM1 patient-derived myotubes and a DM1 mouse model, leaving short repeats of r(CUG) untouched. In contrast, oligonucleotides that recognize r(CUG) sequence rather than structure cleave both long and short r(CUG)-containing transcripts. Transcriptomic, histological, and phenotypic studies demonstrate that Cugamycin broadly and specifically relieves DM1-associated defects in vivo without detectable off-targets. Thus, small molecules that bind and cleave RNA have utility as lead chemical probes and medicines and can selectively target disease-causing RNA structures to broadly improve defects in preclinical animal models.


Subject(s)
Bleomycin/analogs & derivatives , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Oligonucleotides/chemistry , RNA Splicing/genetics , RNA/genetics , RNA/metabolism , Trinucleotide Repeat Expansion/genetics , Animals , Bleomycin/chemistry , Disease Models, Animal , Drug Design , Humans , Mice , Oligonucleotides/metabolism
20.
Mol Pharmacol ; 100(1): 73-82, 2021 07.
Article in English | MEDLINE | ID: mdl-33958481

ABSTRACT

Communication between neuronal cells, which is central to brain function, is performed by several classes of ligand-gated ionotropic receptors. The gold-standard technique for measuring rapid receptor response to agonist is manual patch-clamp electrophysiology, capable of the highest temporal resolution of any current electrophysiology technique. We report an automated high-precision patch-clamp system that substantially improves the throughput of these time-consuming pharmacological experiments. The patcherBotPharma enables recording from cells expressing receptors of interest and manipulation of them to enable millisecond solution exchange to activate ligand-gated ionotropic receptors. The solution-handling control allows for autonomous pharmacological concentration-response experimentation on adherent cells, lifted cells, or excised outside-out patches. The system can perform typical ligand-gated ionotropic receptor experimentation protocols autonomously, possessing a high success rate in completing experiments and up to a 10-fold reduction in research effort over the duration of the experiment. Using it, we could rapidly replicate previous data sets, reducing the time it took to produce an eight-point concentration-response curve of the effect of propofol on GABA type A receptor deactivation from likely weeks of recording to ∼13 hours of recording. On average, the rate of data collection of the patcherBotPharma was a data point every 2.1 minutes that the operator spent interacting with the patcherBotPharma The patcherBotPharma provides the ability to conduct complex and comprehensive experimentation that yields data sets not normally within reach of conventional systems that rely on constant human control. This technical advance can contribute to accelerating the examination of the complex function of ion channels and the pharmacological agents that act on them. SIGNIFICANCE STATEMENT: This work presents an automated intracellular pharmacological electrophysiology robot, patcherBotPharma, that substantially improves throughput and reduces human time requirement in pharmacological patch-clamp experiments. The robotic system includes millisecond fluid exchange handling and can perform highly efficient ligand-gated ionotropic receptor experiments. The patcherBotPharma is built using a conventional patch-clamp rig, and the technical advances shown in this work greatly accelerate the ability to conduct high-fidelity pharmacological electrophysiology.


Subject(s)
Neurons/cytology , Patch-Clamp Techniques/instrumentation , Receptors, GABA-A/metabolism , Animals , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Mice , Neurons/metabolism , Primary Cell Culture , Rats , Robotics
SELECTION OF CITATIONS
SEARCH DETAIL