82 results on '"Eric T. Wang"'
Search Results
2. The myonuclear domain in adult skeletal muscle fibres: past, present and future
- Author
-
James R. Bagley, Lance T. Denes, John J. McCarthy, Eric T. Wang, and Kevin A. Murach
- Subjects
Physiology - Published
- 2023
3. Choroid plexus mis-splicing and altered cerebrospinal fluid composition in myotonic dystrophy type 1
- Author
-
Curtis A Nutter, Benjamin M Kidd, Helmut A Carter, Johanna I Hamel, Philip M Mackie, Nayha Kumbkarni, Mackenzie L Davenport, Dana M Tuyn, Adithya Gopinath, Peter D Creigh, Łukasz J Sznajder, Eric T Wang, Laura P W Ranum, Habibeh Khoshbouei, John W Day, Jacinda B Sampson, Stefan Prokop, and Maurice S Swanson
- Subjects
Neurology (clinical) - 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 knockin 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 knockin 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 donors without neurologically unaffected (two females, three males; ages 50-70) and myotonic dystrophy type 1 donors (one female, three males; ages 50-70). 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) and non-myotonic dystrophy patients (three females, four males; ages 26-51) 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 knockin 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 knockin 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.
- Published
- 2023
4. Mice lacking MBNL1 and MBNL2 exhibit sudden cardiac death and molecular signatures recapitulating myotonic dystrophy
- Author
-
Kuang-Yung Lee, Carol Seah, Ching Li, Yu-Fu Chen, Chwen-Yu Chen, Ching-I Wu, Po-Cheng Liao, Yu-Chiau Shyu, Hailey R Olafson, Kendra K McKee, Eric T Wang, Chi-Hsiao Yeh, and Chao-Hung Wang
- Subjects
Mice, Knockout ,EGF Family of Proteins ,RNA-Binding Proteins ,General Medicine ,DNA-Binding Proteins ,Alternative Splicing ,Mice ,Death, Sudden, Cardiac ,Genetics ,Animals ,Calsequestrin ,Myotonic Dystrophy ,Myocytes, Cardiac ,Muscle, Skeletal ,Molecular Biology ,Genetics (clinical) - 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.
- Published
- 2022
5. The skeletal muscle circadian clock regulates titin splicing through RBM20
- Author
-
Lance A. Riley, Xiping Zhang, Collin M. Douglas, Joseph M. Mijares, David W. Hammers, Christopher A. Wolff, Neil B. Wood, Hailey R. Olafson, Ping Du, Siegfried Labeit, Michael J. Previs, Eric T. Wang, and Karyn A. Esser
- Subjects
RNA Splicing ,Circadian clock ,Immunoglobulin domain ,Biology ,Sarcomere ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Muscular Diseases ,Circadian Clocks ,medicine ,Animals ,Protein Isoforms ,Connectin ,Circadian rhythm ,Muscle, Skeletal ,General Immunology and Microbiology ,Myogenesis ,General Neuroscience ,Skeletal muscle ,RNA-Binding Proteins ,General Medicine ,Cell biology ,Circadian Rhythm ,medicine.anatomical_structure ,RNA splicing ,biology.protein ,Titin ,Protein Kinases - Abstract
Circadian rhythms in skeletal muscle are maintained by a transcriptional-translational feedback loop known as the molecular clock. While previous research suggested a role for the molecular clock in regulating skeletal muscle structure and function, no mechanisms have connected the molecular clock to sarcomeric proteins. Utilizing inducible, skeletal muscle specific, Bmal1 knockout (iMSBmal1-/-) mice, we show that deletion of the skeletal muscle molecular clock alters titin isoform and skeletal muscle sarcomere length. We then use U7 snRNPs in myotubes to directly alter titin splicing in vitro. Truncating titin’s proximal Ig domain results in altered sarcomere length. Finally, we identify a mechanism whereby the skeletal muscle molecular clock regulates titin isoform expression through RBM20, a potent splicing regulator of the titin transcript. Our findings demonstrate the importance of the skeletal muscle molecular clock in maintaining sarcomere length homogeneity through its regulation of RBM20 expression. Because circadian rhythm disruption is a feature of many diseases, our results highlight a pathway that could be targeted to maintain skeletal muscle structure and function in a range of pathologies. Significance Statement Circadian rhythms regulate many aspects of skeletal muscle physiology; however, the exact mechanisms connecting the molecular underpinnings of these rhythms to skeletal muscle structure and function are poorly understood. Here we describe how the skeletal muscle molecular clock regulates titin, the protein ruler regulating sarcomere length and muscle strength. Since circadian rhythms and skeletal muscle weakness underly a number of diseases, these results highlight a potential target for future therapeutic strategies.
- Published
- 2022
6. Author response: The skeletal muscle circadian clock regulates titin splicing through RBM20
- Author
-
Lance A Riley, Xiping Zhang, Collin M Douglas, Joseph M Mijares, David W Hammers, Christopher A Wolff, Neil B Wood, Hailey R Olafson, Ping Du, Siegfried Labeit, Michael J Previs, Eric T Wang, and Karyn A Esser
- Published
- 2022
7. Muscleblind-like proteins use modular domains to localize RNAs by riding kinesins and docking to membranes
- Author
-
Ryan P. Hildebrandt, Kathryn R. Moss, Aleksandra Janusz-Kaminska, Luke A. Knudson, Lance T. Denes, Tanvi Saxena, Devi Prasad Boggupalli, Zhuangyue Li, Kun Lin, Gary J. Bassell, and Eric T. Wang
- Abstract
SummaryRNA transport and local translation provide spatial control of gene expression, and RNA binding proteins (RBPs) act as critical adapters in this multi-step process. Muscleblind-like (MBNL) RNA binding proteins, implicated in myotonic dystrophy and cancer, localize RNAs to myoblast membranes and distal neurites through unknown mechanisms. We found that MBNL forms motile and anchored granules in neurons and myoblasts, and selectively associates with kinesins Kif1bα and Kif1c through its zinc finger (ZnF) domains. Other RBPs with similar ZnFs also associate with these kinesins, implicating a motor-RBP specificity code. Live cell imaging and fractionation revealed that membrane anchoring is mediated through the unstructured carboxy-terminal tail of MBNL1. Both kinesin- and membrane-recruitment functions were reconstituted using MBNL-MS2 coat protein fusions. This approach, termed RBP Module Recruitment and Imaging (RBP-MRI), decouples RNA binding, kinesin recruitment, and membrane anchoring functions, while also establishing general strategies for studying multi-functional, modular domains of RBPs.
- Published
- 2022
8. Repeat length increases disease penetrance and severity in C9orf72 ALS/FTD BAC transgenic mice
- Author
-
Hailey Olafson, Amrutha Pattamatta, Jared I Richardson, J. Andrew Berglund, Tao Zu, Marina M. Scotti, Lauren A. Laboissonniere, Laura P.W. Ranum, Lien Nguyen, and Eric T. Wang
- Subjects
0301 basic medicine ,Transgene ,Mice, Transgenic ,Penetrance ,Biology ,medicine.disease_cause ,Mice ,03 medical and health sciences ,0302 clinical medicine ,C9orf72 ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Genetics (clinical) ,Mutation ,DNA Repeat Expansion ,C9orf72 Protein ,Amyotrophic Lateral Sclerosis ,Alternative splicing ,General Medicine ,Phenotype ,Alternative Splicing ,Disease Models, Animal ,030104 developmental biology ,Frontotemporal Dementia ,General Article ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery - 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.
- Published
- 2020
9. Transcriptome-wide organization of subcellular microenvironments revealed by ATLAS-Seq
- Author
-
Eric T. Wang and Danielle A Adekunle
- Subjects
Cell Extracts ,Sucrose ,AcademicSubjects/SCI00010 ,Intracellular Space ,RNA-binding protein ,Data Resources and Analyses ,Computational biology ,Biology ,Cell Fractionation ,Ribosome ,Mass Spectrometry ,Transcriptome ,Mice ,RNA Isoforms ,Centrifugation, Density Gradient ,Genetics ,Animals ,Coding region ,Narese/9 ,Cellular compartment ,Sequence Analysis, RNA ,RNA-Binding Proteins ,RNA ,Cellular Microenvironment ,Liver ,Proteome ,Female ,Ribosomes - 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.
- Published
- 2020
10. Negative autoregulation mitigates collateral RNase activity of repeat-targeting CRISPR-Cas13d in mammalian cells
- Author
-
Chase P Kelley, Maja C Haerle, and Eric T Wang
- Subjects
Gene Editing ,Mammals ,Ribonucleases ,Animals ,Homeostasis ,Humans ,Myotonic Dystrophy ,RNA ,CRISPR-Cas Systems ,General Biochemistry, Genetics and Molecular Biology ,RNA, Guide, Kinetoplastida - Abstract
Cas13 is a unique family of CRISPR endonucleases exhibiting programmable binding and cleavage of RNAs and is a strong candidate for eukaryotic RNA knockdown in the laboratory and the clinic. However, sequence-specific binding of Cas13 to the target RNA unleashes non-specific bystander RNA cleavage, or collateral activity, which may confound knockdown experiments and raises concerns for therapeutic applications. Although conserved across orthologs and robust in cell-free and bacterial environments, the extent of collateral activity in mammalian cells remains disputed. Here, we investigate Cas13d collateral activity in the context of an RNA-targeting therapy for myotonic dystrophy type 1, a disease caused by a transcribed long CTG repeat expansion. We find that when targeting CUGn RNA in HeLa and other cell lines, Cas13d depletes endogenous and transgenic RNAs, interferes with critical cellular processes, and activates stress response and apoptosis pathways. We also observe collateral effects when targeting other repetitive and unique transgenic sequences, and we provide evidence for collateral activity when targeting highly expressed endogenous transcripts. To minimize collateral activity for repeat-targeting Cas13d therapeutics, we introduce gRNA excision for negative-autoregulatory optimization (GENO), a simple strategy that leverages crRNA processing to control Cas13d expression and is easily integrated into an AAV gene therapy. We argue that thorough assessment of collateral activity is necessary when applying Cas13d in mammalian cells and that implementation of GENO illustrates the advantages of compact and universally robust regulatory systems for Cas-based gene therapies.
- Published
- 2021
11. A comprehensive atlas of fetal splicing patterns in the brain of adult myotonic dystrophy type 1 patients
- Author
-
Max J F Degener, Remco T P van Cruchten, Brittney A Otero, Eric T Wang, Derick G Wansink, and Peter A C ‘t Hoen
- Subjects
musculoskeletal diseases ,Alternative splicing ,Central nervous system ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Biology ,medicine.disease ,Bioinformatics ,Phenotype ,Myotonic dystrophy ,chemistry.chemical_compound ,Exon ,All institutes and research themes of the Radboud University Medical Center ,medicine.anatomical_structure ,chemistry ,RNA splicing ,medicine ,MBNL1 ,splice ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] - Abstract
In patients with myotonic dystrophy type 1 (DM1), dysregulation of RNA-binding proteins like MBNL and CELF1 leads to alternative splicing of exons and is thought to induce a return to fetal splicing patterns in adult tissues, including the central nervous system (CNS). To comprehensively evaluate this, we created an atlas of developmentally regulated splicing patterns in the frontal cortex of healthy individuals and DM1 patients by combining RNA-seq data from BrainSpan, GTEx and DM1 patients. Thirty four splice events displayed an inclusion pattern in DM1 patients that is typical for the fetal situation in healthy individuals. The regulation of DM1-relevant splicing patterns could partly be explained by changes in mRNA expression of the splice regulators MBNL1, MBNL2 and CELF1. On the contrary, interindividual differences in splicing patterns between healthy adults could not be explained by differential expression of these splice regulators. Our findings lend transcriptome-wide evidence to the previously noted shift to fetal splicing patterns in the adult DM1 brain as a consequence of an imbalance in antagonistic MBNL and CELF1 activities. Our atlas serves as a solid foundation for further study and understanding of the cognitive phenotype in patients.
- Published
- 2021
12. Altered Behavioral Responses Show GABA Sensitivity in Muscleblind-Like 2-Deficient Mice: Implications for CNS Symptoms in Myotonic Dystrophy
- Author
-
Kamyra S. Edokpolor, Anwesha Banerjee, Zachary T. McEachin, Jingsheng Gu, Adam Kosti, Juan D. Arboleda, Paul S. García, Eric T. Wang, and Gary J. Bassell
- Subjects
Flumazenil ,Mice, Knockout ,Diazepam ,General Neuroscience ,RNA-Binding Proteins ,General Medicine ,Receptors, GABA-A ,Zolpidem ,Disease Models, Animal ,Mice ,Sevoflurane ,Animals ,Humans ,Myotonic Dystrophy ,RNA ,RNA, Messenger ,gamma-Aminobutyric Acid - Abstract
Considerable evidence from mouse models and human postmortem brain suggests loss of Muscleblind-like protein 2 (MBNL2) function in brain is a major driver of CNS symptoms in Myotonic dystrophy type 1 (DM1). Increased hypersomnia, fatigue, and surgical complications associated with general anesthesia suggest possible sensitivity to GABAergic inhibition in DM1. To test the hypothesis that MBNL2 depletion leads to behavioral sensitivity to GABAAreceptor (GABAA-R) modulation,Mbnl2knock-out (KO) and wild-type (WT) littermates were treated with the anesthetic sevoflurane, the benzodiazepine diazepam, the imidazopyridine zolpidem, and the benzodiazepine rescue agent, flumazenil (Ro 15-1788), and assessed for various behavioral metrics.Mbnl2KO mice exhibited delayed recovery following sevoflurane, delayed emergence and recovery from zolpidem, and enhanced sleep time at baseline that was modulated by flumazenil. A significantly higher proportion ofMbnl2KO mice also loss their righting reflex [loss of righting reflex (LORR)] from a standard diazepam dose. We further examined whether MBNL2 depletion affects total GABAA-R mRNA subunit levels and validated RNA-sequencing data of mis-splicedGabrg2, whose isoform ratios are known to regulate GABA sensitivity and associated behaviors. While no other GABAA-R subunit mRNA levels tested were altered inMbnl2KO mouse prefrontal cortex,Gabrg2S/LmRNA ratio levels were significantly altered. Taken together, our findings indicate that loss of MBNL2 function affects GABAergic function in a mouse model of myotonic dystrophy (DM1).
- Published
- 2022
13. Goals in tension: motivated by genetic disease yet rooted in basic science
- Author
-
Eric T. Wang
- Subjects
Motivation ,DNA Repeat Expansion ,Basic science ,Science ,Translational research ,Neurodegenerative Diseases ,Cell Biology ,Disease ,Connection (mathematics) ,Epistemology ,Translational Research, Biomedical ,Humans ,Psychology ,Molecular Biology ,Biological sciences ,Goals - Abstract
A familial connection to disease can motivate a career in the biological sciences. However, tension may exist over how much effort to devote to fundamental versus translational research. In reality, both pursuits are symbiotic — they synergize to advance our understanding of life and the development of effective therapeutics. Eric Wang’s familial connection to disease raised tension between efforts devoted to fundamental versus translational research — until he realized both synergize.
- Published
- 2021
14. Cell-type-specific dysregulation of RNA alternative splicing in short tandem repeat mouse knockin models of myotonic dystrophy
- Author
-
Jodi L. Bubenik, Helmut A. Carter, Maurice S. Swanson, Ruan Oliveira, Eric T. Wang, Brittney A. Otero, Belinda S. Pinto, Franjo Ivankovic, Curtis A. Nutter, Łukasz J. Sznajder, Eric A. Vitriol, and Benjamin M. Kidd
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,RNA Splicing ,Muscle Fibers, Skeletal ,Biology ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Research Communication ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genome editing ,Genetics ,medicine ,Animals ,Myotonic Dystrophy ,Myocyte ,CRISPR ,Gene Knock-In Techniques ,Muscle, Skeletal ,3' Untranslated Regions ,Gene ,030304 developmental biology ,0303 health sciences ,Alternative splicing ,Gene Expression Regulation, Developmental ,RNA-Binding Proteins ,medicine.disease ,Cell biology ,DNA-Binding Proteins ,Alternative Splicing ,Disease Models, Animal ,030220 oncology & carcinogenesis ,Choroid Plexus ,Mutation ,RNA splicing ,Choroid plexus ,Microsatellite Repeats ,Developmental Biology - 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.
- Published
- 2019
15. A CTG repeat-selective chemical screen identifies microtubule inhibitors as selective modulators of toxic CUG RNA levels
- Author
-
Kaalak Reddy, Jana R. Jenquin, J. Andrew Berglund, Maja C Haerle, Eric T. Wang, Jared I Richardson, Belinda S. Pinto, Lori Planco, John D. Cleary, Ona L. McConnell, Masayuki Nakamori, and Elizabeth Delgado
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Cell ,Drug Evaluation, Preclinical ,Mice, Transgenic ,Biology ,Microtubules ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Small Molecule Libraries ,Mice ,Transcription (biology) ,Microtubule ,Gene expression ,medicine ,Animals ,Humans ,Myotonic Dystrophy ,Gene ,Multidisciplinary ,Alternative splicing ,RNA ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,PNAS Plus ,Trinucleotide Repeat Expansion ,HeLa Cells - 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.
- Published
- 2019
16. Transcriptome alterations in myotonic dystrophy skeletal muscle and heart
- Author
-
John W. Day, Sam Sedehizadeh, Thomas A. Cooper, Charles A. Thornton, Christopher B. Burge, Daniel J. Treacy, Thomas T. Wang, David E. Housman, Katy Eichinger, Amanda J. Ward, J. Andrew Berglund, Adam J. Struck, Kirti Bhatt, Joseph Estabrook, Eric T. Wang, David Brook, Hailey Olafson, and Tony Westbrook
- Subjects
Adult ,Male ,RNA Splicing ,Duchenne muscular dystrophy ,Biology ,Myotonic dystrophy ,Genome ,Transcriptome ,03 medical and health sciences ,Exon ,Gene expression ,Genetics ,medicine ,Humans ,Myotonic Dystrophy ,Muscle, Skeletal ,Molecular Biology ,Genetics (clinical) ,Principal Component Analysis ,0303 health sciences ,Base Sequence ,Gene Expression Profiling ,Myocardium ,030305 genetics & heredity ,RNA-Binding Proteins ,Skeletal muscle ,Heart ,General Medicine ,medicine.disease ,Alternative Splicing ,medicine.anatomical_structure ,RNA splicing ,RNA ,Female ,General Article ,Microsatellite Repeats - Abstract
Myotonic dystrophy (dystrophia myotonica, DM) is a multi-systemic disease caused by expanded CTG or CCTG microsatellite repeats. Characterized by symptoms in muscle, heart and central nervous system, among others, it is one of the most variable diseases known. A major pathogenic event in DM is the sequestration of muscleblind-like proteins by CUG or CCUG repeat-containing RNAs transcribed from expanded repeats, and differences in the extent of MBNL sequestration dependent on repeat length and expression level may account for some portion of the variability. However, many other cellular pathways are reported to be perturbed in DM, and the severity of specific disease symptoms varies among individuals. To help understand this variability and facilitate research into DM, we generated 120 RNASeq transcriptomes from skeletal and heart muscle derived from healthy and DM1 biopsies and autopsies. A limited number of DM2 and Duchenne muscular dystrophy samples were also sequenced. We analyzed splicing and gene expression, identified tissue-specific changes in RNA processing and uncovered transcriptome changes strongly correlating with muscle strength. We created a web resource at http://DMseq.org that hosts raw and processed transcriptome data and provides a lightweight, responsive interface that enables browsing of processed data across the genome.
- Published
- 2018
17. Molecular characterization of myotonic dystrophy fibroblast cell lines for use in small molecule screening
- Author
-
Jana R. Jenquin, Alana P. O’Brien, Kiril Poukalov, Yidan Lu, Jesus A. Frias, Hannah K. Shorrock, Jared I. Richardson, Hormoz Mazdiyasni, Hongfen Yang, Robert W. Huigens, David Boykin, Laura P.W. Ranum, John Douglas Cleary, Eric T. Wang, and J. Andrew Berglund
- Subjects
Multidisciplinary - Abstract
Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are common forms of adult onset muscular dystrophy. Pathogenesis in both diseases is largely driven by production of toxic-expanded repeat RNAs that sequester MBNL RNA-binding proteins, causing mis-splicing. Given this shared pathogenesis, we hypothesized that diamidines, small molecules that rescue mis-splicing in DM1 models, could also rescue mis-splicing in DM2 models. While several DM1 cell models exist, few are available for DM2 limiting research and therapeutic development. Here, we characterize DM1 and DM2 patient-derived fibroblasts for use in small molecule screens and therapeutic studies. We identify mis-splicing events unique to DM2 fibroblasts and common events shared with DM1 fibroblasts. We show that diamidines can partially rescue molecular phenotypes in both DM1 and DM2 fibroblasts. This study demonstrates the potential of fibroblasts as models for DM1 and DM2, which will help meet an important need for well-characterized DM2 cell models.
- Published
- 2021
18. Automated Intracellular Pharmacological Electrophysiology for Ligand-Gated Ionotropic Receptor and Pharmacology Screening
- Author
-
Ona L. McConnell, Mighten C Yip, Andrew Jenkins, Riley E. Perszyk, Craig R. Forest, Stephen F. Traynelis, and Eric T. Wang
- Subjects
Agonist ,Patch-Clamp Techniques ,Computer science ,medicine.drug_class ,Primary Cell Culture ,Drug Evaluation, Preclinical ,CHO Cells ,Mice ,Cricetulus ,medicine ,Animals ,Humans ,Patch clamp ,Receptor ,Ion channel ,Pharmacology ,Neurons ,GABAA receptor ,Robotics ,Articles ,Receptors, GABA-A ,Rats ,Electrophysiology ,HEK293 Cells ,Molecular Medicine ,Ligand-gated ion channel ,Neuroscience ,Ionotropic effect - 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 patcherBot(Pharma) 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 patcherBot(Pharma) was a data point every 2.1 minutes that the operator spent interacting with the patcherBot(Pharma). The patcherBot(Pharma) 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, patcherBot(Pharma), 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 patcherBot(Pharma) 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.
- Published
- 2020
19. Muscleblind-like RNA binding proteins interact with kinesin motors to localize mRNA in neurons but impairments occur in an in vitro model of myotonic dystrophy DM1
- Author
-
Eric T. Wang and Gary J. Bassell
- Published
- 2020
20. Small-molecule targeted recruitment of a nuclease to cleave an oncogenic RNA in a mouse model of metastatic cancer
- Author
-
Jessica L. Childs-Disney, Yue Li, Ilyas Yildirim, Alexander Adibekian, Yoshio Nakai, Haruo Aikawa, Eric T. Wang, Matthew D. Disney, Tanya Khan, Matthew G. Costales, Daniel Abegg, Sai Pradeep Velagapudi, Dominic Gregor Hoch, and Kye Won Wang
- Subjects
Chemical biology ,Oligonucleotides ,Breast Neoplasms ,01 natural sciences ,Transcriptome ,Small Molecule Libraries ,03 medical and health sciences ,Mice ,Ribonucleases ,Cell Line, Tumor ,Animals ,Humans ,Ribonuclease ,Neoplasm Metastasis ,030304 developmental biology ,0303 health sciences ,Nuclease ,Multidisciplinary ,biology ,Molecular Structure ,010405 organic chemistry ,Oligonucleotide ,Chemistry ,RNA ,Small molecule ,0104 chemical sciences ,3. Good health ,Cell biology ,Disease Models, Animal ,MicroRNAs ,Drug Design ,biology.protein ,Nucleic acid ,Female - 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.
- Published
- 2020
21. Microtubule-based transport is essential to distribute RNA and nascent protein in skeletal muscle
- Author
-
Chase P. Kelley, Lance T. Denes, and Eric T. Wang
- Subjects
Sarcomeres ,RNA localization ,Cellular differentiation ,Science ,Muscle Fibers, Skeletal ,Neuromuscular Junction ,Biophysics ,General Physics and Astronomy ,RNA transport ,Biology ,Muscle Development ,Ribosome ,Sarcomere ,Microtubules ,General Biochemistry, Genetics and Molecular Biology ,Article ,Polymerization ,Mice ,Microtubule ,Protein biosynthesis ,medicine ,Animals ,Computer Simulation ,RNA, Messenger ,Muscle, Skeletal ,Regulation of gene expression ,Cell Nucleus ,Syncytium ,Multidisciplinary ,Chemistry ,Nocodazole ,Skeletal muscle ,RNA ,Biological Transport ,Cell Differentiation ,General Chemistry ,Cytoskeletal Filaments ,Translocon ,Cell biology ,Molecular Imaging ,medicine.anatomical_structure ,Ribonucleoproteins ,Protein Biosynthesis ,Differentiation ,Ribosomes - Abstract
While the importance of RNA localization in highly differentiated cells is well appreciated, basic principles of RNA localization in skeletal muscle remain poorly characterized. Here, we develop a method to detect and quantify single molecule RNA localization patterns in skeletal myofibers, and uncover a critical role for directed transport of RNPs in muscle. We find that RNAs localize and are translated along sarcomere Z-disks, dispersing tens of microns from progenitor nuclei, regardless of encoded protein function. We find that directed transport along the lattice-like microtubule network of myofibers becomes essential to achieve this localization pattern as muscle development progresses; disruption of this network leads to extreme accumulation of RNPs and nascent protein around myonuclei. Our observations suggest that global active RNP transport may be required to distribute RNAs in highly differentiated cells and reveal fundamental mechanisms of gene regulation, with consequences for myopathies caused by perturbations to RNPs or microtubules., It is increasingly recognised that the spatial localisation of RNA is important for proper cellular function. Here, the authors investigate RNA localisation in skeletal muscle and develop methods to show that global active transport of RNA is required to maintain dispersion of gene products in the large muscle syncytium.
- Published
- 2022
22. Author Correction: Molecular mechanisms underlying nucleotide repeat expansion disorders
- Author
-
Peter K. Todd, Indranil Malik, Chase P. Kelley, and Eric T. Wang
- Subjects
chemistry.chemical_classification ,Text mining ,chemistry ,business.industry ,Nucleotide ,Cell Biology ,Computational biology ,Trinucleotide repeat expansion ,business ,Molecular Biology - Published
- 2021
23. Aberrant Myokine Signaling in Congenital Myotonic Dystrophy
- Author
-
Maurice S. Swanson, Masanori P. Takahashi, Hideki Mochizuki, Ichizo Nishino, Yukiko K. Hayashi, James D. Thomas, Masayuki Nakamori, Eric T. Wang, and Kohei Hamanaka
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,CCCTC-Binding Factor ,medicine.medical_specialty ,Transcription, Genetic ,Muscle Proteins ,Biology ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,Myokine ,medicine ,Humans ,Myotonic Dystrophy ,Myocyte ,lcsh:QH301-705.5 ,Ataxin-7 ,Binding Sites ,Interleukin-6 ,Muscles ,Infant ,RNA-Binding Proteins ,DNA Methylation ,medicine.disease ,Phenotype ,Muscle atrophy ,Up-Regulation ,Alternative Splicing ,030104 developmental biology ,Endocrinology ,lcsh:Biology (General) ,DNA methylation ,CpG Islands ,medicine.symptom ,Signal transduction ,Trinucleotide Repeat Expansion ,030217 neurology & neurosurgery ,Protein Binding ,Signal Transduction - Abstract
Summary: Myotonic dystrophy types 1 (DM1) and 2 (DM2) are dominantly inherited neuromuscular disorders caused by a toxic gain of function of expanded CUG and CCUG repeats, respectively. Although both disorders are clinically similar, congenital myotonic dystrophy (CDM), a severe DM form, is found only in DM1. CDM is also characterized by muscle fiber immaturity not observed in adult DM, suggesting specific pathological mechanisms. Here, we revealed upregulation of the interleukin-6 (IL-6) myokine signaling pathway in CDM muscles. We also found a correlation between muscle immaturity and not only IL-6 expression but also expanded CTG repeat length and CpG methylation status upstream of the repeats. Aberrant CpG methylation was associated with transcriptional dysregulation at the repeat locus, increasing the toxic RNA burden that upregulates IL-6. Because the IL-6 pathway is involved in myocyte maturation and muscle atrophy, our results indicate that enhanced RNA toxicity contributes to severe CDM phenotypes through aberrant IL-6 signaling. : Congenital myotonic dystrophy (CDM) manifests characteristic genetic (very large CTG repeat expansions), epigenetic (CpG hypermethylation upstream of the repeat), and phenotypic (muscle immaturity) features not seen in adult DM. Nakamori et al. find phenotype-genotype and epigenotype correlation in CDM muscle and reveal involvement of the IL-6 myokine signaling pathway in the disease process. Keywords: CTCF, ER stress, IL-6, muscular dystrophy, NF-κB, trinucleotide, cytokine, splicing
- Published
- 2017
24. Myotonic dystrophy: approach to therapy
- Author
-
Charles A. Thornton, Eric T. Wang, and Ellie M. Carrell
- Subjects
0301 basic medicine ,RNA-binding protein ,Biology ,Bioinformatics ,Myotonic dystrophy ,Article ,Myotonin-Protein Kinase ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Myotonic Dystrophy ,RNA, Antisense ,Dominance (genetics) ,Regulation of gene expression ,Myotonin-protein kinase ,Genetic disorder ,RNA-Binding Proteins ,RNA ,Genetic Therapy ,medicine.disease ,030104 developmental biology ,Gene Expression Regulation ,Mutation ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,Developmental Biology - Abstract
Myotonic dystrophy (DM) is a dominantly-inherited genetic disorder affecting skeletal muscle, heart, brain, and other organs. DM type 1 is caused by expansion of a CTG triplet repeat in DMPK, whereas DM type 2 is caused by expansion of a CCTG tetramer repeat in CNBP. In both cases the DM mutations lead to expression of dominant-acting RNAs. Studies of RNA toxicity have now revealed novel mechanisms and new therapeutic targets. Preclinical data have suggested that RNA dominance is responsive to therapeutic intervention and that DM therapy can be approached at several different levels. Here we review recent efforts to alleviate RNA toxicity in DM.
- Published
- 2017
25. Myotonic dystrophy: disease repeat range, penetrance, age of onset, and relationship between repeat size and phenotypes
- Author
-
Kevin Yum, Eric T. Wang, and Auinash Kalsotra
- Subjects
0301 basic medicine ,Neuromuscular disease ,Penetrance ,Disease ,Biology ,Myotonic dystrophy ,Article ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Myotonic Dystrophy ,Genetic Testing ,Age of Onset ,Repetitive Sequences, Nucleic Acid ,Genetic testing ,medicine.diagnostic_test ,medicine.disease ,Myotonia ,Phenotype ,030104 developmental biology ,Microsatellite ,Age of onset ,Developmental Biology - Abstract
Myotonic dystrophy (DM) is an autosomal dominant neuromuscular disease primarily characterized by myotonia and progressive muscle weakness. The pathogenesis of DM involves microsatellite expansions in noncoding regions of transcripts that result in toxic RNA gain-of-function. Each successive generation of DM families carries larger repeat expansions, leading to an earlier age of onset with increasing disease severity. At present, diagnosis of DM is challenging and requires special genetic testing to account for somatic mosaicism and meiotic instability. While progress in genetic testing has been made, more rapid, accurate, and cost-effective approaches for measuring repeat lengths are needed to establish clear correlations between repeat size and disease phenotypes.
- Published
- 2017
26. A Toxic RNA Catalyzes the Cellular Synthesis of Its Own Inhibitor, Shunting It to Endogenous Decay Pathways
- Author
-
Raphael I. Benhamou, Matthew D. Disney, Eric T. Wang, and Alicia J. Angelbello
- Subjects
Clinical Biochemistry ,Chemical biology ,Plasma protein binding ,Biology ,010402 general chemistry ,Biochemistry ,Myotonic dystrophy ,01 natural sciences ,Article ,Cell Line ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,medicine ,RNA Precursors ,Humans ,Myotonic Dystrophy ,MBNL1 ,RNA, Small Interfering ,Molecular Biology ,030304 developmental biology ,Pharmacology ,Zinc finger ,0303 health sciences ,Messenger RNA ,010405 organic chemistry ,Chemistry ,Intron ,RNA-Binding Proteins ,RNA ,Fibroblasts ,medicine.disease ,Small molecule ,Introns ,0104 chemical sciences ,Cell biology ,Alternative Splicing ,Tandem Repeat Sequences ,RNA splicing ,Nucleic acid ,Molecular Medicine ,RNA Interference - Abstract
SUMMARYMyotonic dystrophy type 2 (DM2) is a genetically defined muscular dystrophy caused by a toxic expanded repeat of r(CCUG) [heretofore (CCUG)exp], harbored in intron 1 of CHC-Type Zinc Finger Nucleic Acid Binding Protein (CNBP) pre-mRNA. This r(CCUG)exp causes DM2 via a gain-of-function mechanism that results in three hallmarks of its pathology: (i) binding to RNA-binding proteins (RBPs) that aggregate into nuclear foci; (ii) sequestration of muscleblind-like-1 (MBNL1) protein, a regulator of alternative pre-mRNA splicing, leading to splicing defects; and (iii) retention of intron 1 in the CNBP mRNA. Here, we find that CNBP intron retention is caused by the r(CCUG)exp-MBNL1 complex and can be rescued by small molecules. We studied two types of small molecules with different modes of action, ones that simply bind and ones that can be synthesized by a r(CCUG)exp-templated reaction in cells, that is the RNA synthesizes its own drug. Indeed, our studies completed in DM2 patient-derived fibroblasts show that the compounds disrupt the r(CCUG)exp-MBNL1 complex, reduce intron retention, subjecting the liberated intronic r(CCUG)exp to native decay pathways, and rescue other DM2-associated cellular defects. Collectively, this study shows that small molecules can affect RNA biology by shunting toxic transcripts towards native decay pathways.HIGHLIGHTSIntron retention in RNA repeat expansions can be due to repeats binding to proteinsSmall molecules that bind RNA repeats and inhibit protein binding can trigger decayA toxic RNA repeat can catalyze the synthesis of its own inhibitor on-siteOn-site drug synthesis most potently affects disease biologyeTOC BLURBThe most common way to target RNA is to use antisense oligonucleotides to target unstructured RNAs for destruction. Here, we show for the first time that small molecules targeting structured, disease-causing RNAs can shunt them towards native decay pathways by affecting their processing.
- Published
- 2019
27. Sleep disorders in myotonic dystrophies
- Author
-
Eric T. Wang, James Wymer, S. H. Subramony, and Belinda S. Pinto
- Subjects
0301 basic medicine ,Sleep Wake Disorders ,Pediatrics ,medicine.medical_specialty ,Movement disorders ,Physiology ,Excessive daytime sleepiness ,030105 genetics & heredity ,Myotonic dystrophy ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Physiology (medical) ,medicine ,Humans ,Myotonic Dystrophy ,Fatigue ,business.industry ,Sleep control ,Sleep apnea ,medicine.disease ,Sleep in non-human animals ,Poor sleep ,Neurology (clinical) ,medicine.symptom ,Abnormality ,business ,030217 neurology & neurosurgery - Abstract
Myotonic dystrophies (DM), the most common muscular dystrophies, are known to have significant sleep disturbances. We analyzed the literature on sleep and excessive daytime sleepiness (EDS) in DM over the past 30 years. In this review we provide a brief overview of sleep, sleep disorders, and methods of assessment. We also analyze data regarding major sleep disorders in DM patients, including: sleep-disordered breathing (SDB), with both central and obstructive sleep apneas (CSA,OSA); EDS; sleep-related movement disorders; and poor sleep quality. We review the possible pathogenesis of these disorders and outline management strategies. We also consider possible future avenues for research. The findings highlight the complex set of sleep-related problems, including the primary abnormality of sleep control in myotonic dystrophies. In individual patients the roles of poor sleep hygiene, SDB, primary hypersomnia, and excess fatigue require careful assessment for appropriate management.
- Published
- 2019
28. Culturing C2C12 myotubes on micromolded gelatin hydrogels accelerates myotube maturation
- Author
-
Karyn A. Esser, Joseph R. Mijares, Kendra K. McKee, Lance A. Riley, Juan Arboleda, Lance T. Denes, and Eric T. Wang
- Subjects
0301 basic medicine ,Myofilament ,lcsh:Diseases of the musculoskeletal system ,food.ingredient ,Surface Properties ,Myoblasts, Skeletal ,Muscle Fibers, Skeletal ,Cell Culture Techniques ,Muscle Development ,Sarcomere ,Gelatin ,Micromolding ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,food ,medicine ,Animals ,Myocyte ,Orthopedics and Sports Medicine ,RNA-Seq ,Molecular Biology ,Myogenesis ,Research ,Gene Expression Profiling ,Skeletal muscle ,Cell Differentiation ,Hydrogels ,Cell Biology ,RNAseq ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Myotubes ,Self-healing hydrogels ,C2C12 ,lcsh:RC925-935 ,030217 neurology & neurosurgery - Abstract
Background Skeletal muscle contributes to roughly 40% of lean body mass, and its loss contributes to morbidity and mortality in a variety of pathogenic conditions. Significant insights into muscle function have been made using cultured cells, in particular, the C2C12 myoblast line. However, differentiation of these cells in vitro typically yields immature myotubes relative to skeletal muscles in vivo. While many efforts have attempted to improve the maturity of cultured myotubes, including the use of bioengineered substrates, lack of molecular characterization has precluded their widespread implementation. This study characterizes morphological, molecular, and transcriptional features of C2C12 myotubes cultured on crosslinked, micropatterned gelatin substrates fabricated using previously established methods and compares them to myotubes grown on unpatterned gelatin or traditional plasticware. Methods We used immunocytochemistry, SDS-PAGE, and RNAseq to characterize C2C12 myotubes grown on micropatterned gelatin hydrogels, unpatterned gelatin hydrogels, and typical cell culture substrates (i.e., plastic or collagen-coated glass) across a differentiation time course. The ability to form aligned sarcomeres and myofilament protein concentration was assessed. Additionally, the transcriptome was analyzed across the differentiation time course. Results C2C12 myotubes grown on micropatterned gelatin hydrogels display an increased ability to form aligned sarcomeres as well as increased contractile protein content relative to myotubes cultured on unpatterned gelatin and plastic. Additionally, genes related to sarcomere formation and in vivo muscle maturation are upregulated in myotubes grown on micropatterned gelatin hydrogels relative to control myotubes. Conclusions Our results suggest that growing C2C12 myotubes on micropatterned gelatin hydrogels accelerates sarcomere formation and yields a more fully matured myotube culture. Thus, the use of micropatterned hydrogels is a viable and simple approach to better model skeletal muscle biology in vitro. Electronic supplementary material The online version of this article (10.1186/s13395-019-0203-4) contains supplementary material, which is available to authorized users.
- Published
- 2019
29. Precise small-molecule cleavage of an r(CUG) repeat expansion in a myotonic dystrophy mouse model
- Author
-
Alicia J. Angelbello, Jonathan L. Chen, Suzanne G. Rzuczek, Walter N. Moss, Michael D. Cameron, Kendra K. McKee, Hailey Olafson, Eric T. Wang, and Matthew D. Disney
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,RNA splicing ,Oligonucleotides ,chemical biology ,01 natural sciences ,Myotonic dystrophy ,03 medical and health sciences ,chemistry.chemical_compound ,Bleomycin ,Mice ,genetic disease ,medicine ,MBNL1 ,CRISPR ,Animals ,Humans ,Myotonic Dystrophy ,Multidisciplinary ,010405 organic chemistry ,Oligonucleotide ,RNA ,Biological Sciences ,medicine.disease ,Small molecule ,3. Good health ,0104 chemical sciences ,Cell biology ,Disease Models, Animal ,nucleic acids ,030104 developmental biology ,chemistry ,Drug Design ,Applied Biological Sciences ,Trinucleotide repeat expansion ,Trinucleotide Repeat Expansion - Abstract
Significance Development of small-molecule lead medicines that potently and specifically modulate RNA function is challenging. We designed a small molecule that cleaves r(CUG)exp, the RNA repeat expansion that causes myotonic dystrophy type 1. In cells and in an animal model, the small-molecule cleaver specifically recognizes the 3-dimensional structure of r(CUG)exp, cleaving it more selectively among transcripts containing short, nonpathogenic r(CUG) repeats than an oligonucleotide that recognizes RNA sequence via Watson-Crick base pairing. The small molecule broadly relieves disease-associated phenotype in a mouse model. Thus, small molecules that recognize and cleave RNA structures should be considered a therapeutic strategy for targeting RNA in vivo., 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.
- Published
- 2019
30. Dysregulation of mRNA Localization and Translation in Genetic Disease
- Author
-
Christina Gross, Ji Ann Lee, Indulekha P. Sudhakaran, Wilfried Rossoll, Kathryn R. Moss, Eric T. Wang, Gary J. Bassell, J. Matthew Taliaferro, Massachusetts Institute of Technology. Department of Biology, and Taliaferro, Jefferson Matthew
- Subjects
0301 basic medicine ,Regulation of gene expression ,General Neuroscience ,Symposium and Mini-Symposium ,RNA-Binding Proteins ,RNA-binding protein ,Translation (biology) ,Spinal muscular atrophy ,Disease ,Biology ,medicine.disease ,Myotonic dystrophy ,Fragile X syndrome ,03 medical and health sciences ,030104 developmental biology ,Gene Expression Regulation ,Fragile X Syndrome ,Synaptic plasticity ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,RNA, Messenger ,Nervous System Diseases ,Neuroscience - Abstract
RNA-binding proteins (RBPs) acting at various steps in the post-transcriptional regulation of gene expression play crucial roles in neuronal development and synaptic plasticity. Genetic mutations affecting several RBPs and associated factors lead to diverse neurological symptoms, as characterized by neurodevelopmental and neuropsychiatric disorders, neuromuscular and neurodegenerative diseases, and can often be multisystemic diseases. We will highlight the physiological roles of a few specific proteins in molecular mechanisms of cytoplasmic mRNA regulation, and how these processes are dysregulated in genetic disease. Recent advances in computational biology and genomewide analysis, integrated with diverse experimental approaches and model systems, have provided new insights into conserved mechanisms and the shared pathobiology of mRNA dysregulation in disease. Progress has been made to understand the pathobiology of disease mechanisms for myotonic dystrophy, spinal muscular atrophy, and fragile X syndrome, with broader implications for other RBP-associated genetic neurological diseases. This gained knowledge of underlying basic mechanisms has paved the way to the development of therapeutic strategies targeting disease mechanisms.
- Published
- 2016
31. Conservation of context-dependent splicing activity in distant Muscleblind homologs
- Author
-
Ona L. McConnell, Tanvi Saxena, Eric T. Wang, J. Andrew Berglund, Julia C. Oddo, Koch Institute for Integrative Cancer Research at MIT, Oddo, Julia Christine, and Wang, Eric T
- Subjects
0301 basic medicine ,RNA Splicing ,Exonic splicing enhancer ,RNA-binding protein ,Biology ,Conserved sequence ,Evolution, Molecular ,Mice ,03 medical and health sciences ,Exon ,Genes, Reporter ,Genetics ,Animals ,Humans ,Placozoa ,RNA, Messenger ,Nucleotide Motifs ,Molecular Biology ,Conserved Sequence ,Repetitive Sequences, Nucleic Acid ,Sequence Homology, Amino Acid ,030102 biochemistry & molecular biology ,Alternative splicing ,Intron ,RNA-Binding Proteins ,RNA ,Exons ,bacterial infections and mycoses ,Introns ,Ciona intestinalis ,3. Good health ,Gene Ontology ,030104 developmental biology ,RNA splicing ,HeLa Cells - Abstract
The Muscleblind (MBL) protein family is a deeply conserved family of RNA binding proteins that regulate alternative splicing, alternative polyadenylation, RNA stability and RNA localization. Their inactivation due to sequestration by expanded CUG repeats causes symptoms in the neuromuscular disease myotonic dystrophy. MBL zinc fingers are the most highly conserved portion of these proteins, and directly interact with RNA. We identified putative MBL homologs in Ciona intestinalis and Trichoplax adhaerens, and investigated their ability, as well as that of MBL homologs from human/mouse, fly and worm, to regulate alternative splicing. We found that all homologs can regulate alternative splicing in mouse cells, with some regulating over 100 events. The cis-elements through which each homolog exerts its splicing activities are likely to be highly similar to mammalian Muscleblind-like proteins (MBNLs), as suggested by motif analyses and the ability of expanded CUG repeats to inactivate homolog-mediated splicing. While regulation of specific target exons by MBL/MBNL has not been broadly conserved across these species, genes enriched for MBL/MBNL binding sites in their introns may play roles in cell adhesion, ion transport and axon guidance, among other biological pathways, suggesting a specific, conserved role for these proteins across a broad range of metazoan species., National Institutes of Health (U.S.) (DP5 OD017865)
- Published
- 2016
32. Identification of new branch points and unconventional introns in Saccharomyces cerevisiae
- Author
-
Boris Zinshteyn, Genevieve M. Gould, Joseph M. Paggi, Christopher B. Burge, Yuchun Guo, David V. Phizicky, David K. Gifford, Wendy V. Gilbert, Eric T. Wang, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology. Department of Biology, Gould, Genevieve Michelle, Paggi, Joseph M., Guo, Yuchun, Phizicky, David Vincent, Zinshteyn, Boris, Wang, Eric T, Gilbert, Wendy, Gifford, David K, and Burge, Christopher B
- Subjects
0301 basic medicine ,RNA Stability ,biology ,Sequence Analysis, RNA ,RNA Splicing ,Saccharomyces cerevisiae ,Intron ,RNA ,Computational biology ,biology.organism_classification ,Introns ,Article ,Yeast ,03 medical and health sciences ,030104 developmental biology ,Meiosis ,RNA splicing ,splice ,RNA Splice Sites ,Nucleotide Motifs ,Molecular Biology - Abstract
Spliced messages constitute one-fourth of expressed mRNAs in the yeast Saccharomyces cerevisiae, and most mRNAs in metazoans. Splicing requires 5′ splice site (5′SS), branch point (BP), and 3′ splice site (3′SS) elements, but the role of the BP in splicing control is poorly understood because BP identification remains difficult. We developed a high-throughput method, Branch-seq, to map BPs and 5′SSs of isolated RNA lariats. Applied to S. cerevisiae, Branch-seq detected 76% of expressed, annotated BPs and identified a comparable number of novel BPs. We performed RNA-seq to confirm associated 3′SS locations, identifying some 200 novel splice junctions, including an AT-AC intron. We show that several yeast introns use two or even three different BPs, with effects on 3′SS choice, protein coding potential, or RNA stability, and identify novel introns whose splicing changes during meiosis or in response to stress. Together, these findings show unanticipated complexity of splicing in yeast., National Science Foundation (U.S.) (Grant 0821391)
- Published
- 2016
33. Identifying Robertsonian Translocation Carriers by Microarray-Based DNA Analysis
- Author
-
Jacob Zahn, Stephanie Huang, Kelli A Davies, Christopher Kingsley, Patrick E. Bogard, Kypros H. Nicolaides, Kara Juneau, Arnold Oliphant, Craig A. Struble, and Eric T Wang
- Subjects
Adult ,0301 basic medicine ,Heterozygote ,Embryology ,Microarray ,Robertsonian translocation ,Trisomy ,Chromosomal translocation ,Prenatal diagnosis ,030105 genetics & heredity ,medicine.disease_cause ,Translocation, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Prenatal Diagnosis ,Centromere ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Oligonucleotide Array Sequence Analysis ,Genetics ,030219 obstetrics & reproductive medicine ,business.industry ,Genetic Carrier Screening ,Obstetrics and Gynecology ,General Medicine ,medicine.disease ,Cell-free fetal DNA ,Pediatrics, Perinatology and Child Health ,Female ,DNA microarray ,business ,Algorithms - Abstract
Objective: To develop a noninvasive prenatal testing improvement that allows identification of Robertsonian translocation carriers. Methods: Blood samples from 191 subjects, including 7 pregnant and 9 non-pregnant Robertsonian translocation carriers, were analyzed for fetal trisomy and Robertsonian translocation status. Digital Analysis of Selected Regions (DANSR™) assays targeting sequences common to the p arms of 5 acrocentric chromosomes were developed and added to existing DANSR assays. DANSR products were hybridized onto a custom DNA microarray for DNA analysis. The Fetal-Fraction Optimized Risk of Trisomy Evaluation (FORTE™) algorithm measures the fraction of fetal DNA and accounts for both the fetal and maternal fractions in the cell-free DNA sample to determine Robertsonian risk. The expectation in a Robertsonian translocation carrier is that DANSR assays on acrocentric p arms should have a concentration 20% less than that of controls. Results: The FORTE algorithm correctly classified the fetal trisomy status and maternal Robertsonian translocation status in all 191 samples. Sixteen samples had a Robertsonian risk score above 99%, while 175 samples had a Robertsonian risk score below 0.01%. Conclusions: Robertsonian translocations are the most common chromosomal translocations and can have significant reproductive consequences. A maternal screen for Robertsonian translocation carriers would provide women valuable information regarding the risk of fetal trisomy.
- Published
- 2016
34. Mice with endogenous <scp>TDP</scp> ‐43 mutations exhibit gain of splicing function and characteristics of amyotrophic lateral sclerosis
- Author
-
David E. Housman, Prasanth Sivakumar, Martina Hallegger, Cristian Bodo, Bernadett Kalmar, Warren Emmett, Hugo Alexandre Mendes Oliveira, Philip Stanier, Adrian M. Isaacs, Alexander E. Conicella, Linda Greensmith, Lydia Teboul, Pietro Fratta, Alessandro Marrero-Gagliardi, Vincent Plagnol, Nicolas L. Fawzi, José M. Brito-Armas, Nicol Birsa, Yichao Yu, Erwin Pauws, Emma Peskett, Joffrey Mianné, Agnieszka M. Ule, Gemma F. Codner, T. Ricketts, Andrea Calvo, Silvia Corrochano, Toby Collins, Jack Humphrey, M Groves, Mark F. Lythgoe, Emanuele Buratti, Francisco E. Baralle, Eric T. Wang, Adriano Chiò, Alan Mejia Maza, Michelle Stewart, Yoichi Gondo, Ryutaro Fukumura, Kitty Lo, Elizabeth M. C. Fisher, Abraham Acevedo-Arozena, Massachusetts Institute of Technology. Department of Biology, Wang, Eric T, and Housman, David E
- Subjects
Genetics and Molecular Biology (all) ,0301 basic medicine ,TDP-43 ,Immunology and Microbiology (all) ,RNA-binding protein ,medicine.disease_cause ,Biochemistry ,Mice ,Exon ,0302 clinical medicine ,Molecular Biology of Disease ,News & Views ,Motor Neurons ,Mutation ,General Neuroscience ,RNA-Binding Proteins ,ALS ,cryptic exon ,skiptic exon ,splicing ,Neuroscience (all) ,Molecular Biology ,Biochemistry, Genetics and Molecular Biology (all) ,Articles ,Exons ,RNA Biology ,Cell biology ,DNA-Binding Proteins ,RNA splicing ,RNA Splicing ,Biology ,TARDBP ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,mental disorders ,medicine ,Animals ,Humans ,Loss function ,General Immunology and Microbiology ,Point mutation ,Amyotrophic Lateral Sclerosis ,Alternative splicing ,nutritional and metabolic diseases ,nervous system diseases ,TDP‐43 ,Alternative Splicing ,030104 developmental biology ,Gene Expression Regulation ,030217 neurology & neurosurgery ,Neuroscience - Abstract
TDP-43 (encoded by the gene TARDBP) is an RNA binding protein central to the pathogenesis of amyotrophic lateral sclerosis (ALS). However, how TARDBP mutations trigger pathogenesis remains unknown. Here, we use novel mouse mutants carrying point mutations in endogenous Tardbp to dissect TDP-43 function at physiological levels both in vitro and in vivo. Interestingly, we find that mutations within the C-terminal domain of TDP-43 lead to a gain of splicing function. Using two different strains, we are able to separate TDP-43 loss- and gain-of-function effects. TDP-43 gain-of-function effects in these mice reveal a novel category of splicing events controlled by TDP-43, referred to as "skiptic" exons, in which skipping of constitutive exons causes changes in gene expression. In vivo, this gain-of-function mutation in endogenous Tardbp causes an adult-onset neuromuscular phenotype accompanied by motor neuron loss and neurodegenerative changes. Furthermore, we have validated the splicing gain-of-function and skiptic exons in ALS patient-derived cells. Our findings provide a novel pathogenic mechanism and highlight how TDP-43 gain of function and loss of function affect RNA processing differently, suggesting they may act at different disease stages. Keywords: ALS; cryptic exon; skiptic exon; splicing; TDP-43
- Published
- 2018
35. A Requirement for Mena, an Actin Regulator, in Local mRNA Translation in Developing Neurons
- Author
-
Christopher B. Burge, Marina Vidaki, Matthew J. Taliaferro, Frauke Drees, Erwin Lanslots, Tanvi Saxena, Frank B. Gertler, Antonios Tatarakis, Eric T. Wang, Massachusetts Institute of Technology. Department of Biology, Koch Institute for Integrative Cancer Research at MIT, Vidaki, Marina, Drees, Frauke, Saxena, Tanvi, Lanslots, Erwin, Taliaferro, Matthew J., Burge, Christopher B, Wang, Eric T, and Gertler, Frank
- Subjects
0301 basic medicine ,DYRK1A ,Neurogenesis ,Synaptogenesis ,Regulator ,macromolecular substances ,Biology ,Article ,03 medical and health sciences ,Cell Movement ,Animals ,RNA, Messenger ,Growth cone ,Ribonucleoprotein ,Neurons ,Regulation of gene expression ,General Neuroscience ,Translation (biology) ,Actins ,Axons ,Cytoskeletal Proteins ,030104 developmental biology ,Gene Expression Regulation ,nervous system ,Protein Biosynthesis ,Axon guidance ,Neuroscience - Abstract
During neuronal development, local mRNA translation is required for axon guidance and synaptogenesis, and dysregulation of this process contributes to multiple neurodevelopmental and cognitive disorders. However, regulation of local protein synthesis in developing axons remains poorly understood. Here, we uncover a novel role for the actin-regulatory protein Mena in the formation of a ribonucleoprotein complex that involves the RNA-binding proteins HnrnpK and PCBP1 and regulates local translation of specific mRNAs in developing axons. We find that translation of dyrk1a, a Down syndrome- and autism spectrum disorders-related gene, is dependent on Mena, both in steady-state conditions and upon BDNF stimulation. We identify hundreds of additional mRNAs that associate with the Mena complex, suggesting that it plays broader role(s) in post-transcriptional gene regulation. Our work establishes a dual role for Mena in neurons, providing a potential link between regulation of actin dynamics and local translation. Keywords: Mena; ENAH; Ena/VASP; developing axon; growth cone; axon guidance; local translation; ribonucleoprotein; Dyrk1a, National Institutes of Health (U.S.) (Grant U01-CA184897), National Cancer Institute (U.S.) (Grant P30-CA14051)
- Published
- 2017
36. Disrupted prenatal RNA processing and myogenesis in congenital myotonic dystrophy
- Author
-
James D. Thomas, Faaiq N. Aslam, Masayuki Nakamori, Zacharias P. Anastasiadis, Eric T. Wang, Maurice S. Swanson, Olgert Bardhi, Łukasz J. Sznajder, Marina M. Scotti, and Ichizo Nishino
- Subjects
0301 basic medicine ,RNA Splicing ,Biology ,Muscle Development ,Myotonic dystrophy ,03 medical and health sciences ,Exon ,chemistry.chemical_compound ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Gene expression ,Genetics ,medicine ,MBNL1 ,Animals ,Humans ,Myotonic Dystrophy ,RNA Processing, Post-Transcriptional ,Cells, Cultured ,Regulation of gene expression ,Myogenesis ,Gene Expression Profiling ,Alternative splicing ,RNA ,Gene Expression Regulation, Developmental ,Infant ,RNA-Binding Proteins ,medicine.disease ,Cell biology ,DNA-Binding Proteins ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Child, Preschool ,Carrier Proteins ,030217 neurology & neurosurgery ,Outlook ,Developmental Biology - 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.
- Published
- 2017
37. Impeding Transcription of Expanded Microsatellite Repeats by Deactivated Cas9
- Author
-
Héctor R. Méndez-Gómez, Ona L. McConnell, Tanvi Saxena, Maurice S. Swanson, Lance T. Denes, Guangbin Xia, Ruan Oliveira, Eric T. Wang, Juan Arboleda, John D. Cleary, and Belinda S. Pinto
- Subjects
0301 basic medicine ,Male ,Transcription, Genetic ,CRISPR-Associated Proteins ,RNA polymerase II ,Myoblasts ,0302 clinical medicine ,Transcription (biology) ,Transduction, Genetic ,CRISPR ,Myotonic Dystrophy ,Guide RNA ,0303 health sciences ,Dependovirus ,Cell biology ,Microsatellite ,Female ,RNA, Guide, Kinetoplastida ,Genetic Vectors ,Down-Regulation ,Mice, Transgenic ,Biology ,Myotonic dystrophy ,Virus ,Article ,03 medical and health sciences ,Chloride Channels ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,C9orf72 Protein ,Cas9 ,RNA ,CD24 Antigen ,Cell Biology ,Genetic Therapy ,Myotonia ,medicine.disease ,Endonucleases ,Molecular biology ,Enzyme Activation ,Alternative Splicing ,Disease Models, Animal ,030104 developmental biology ,HEK293 Cells ,ran GTP-Binding Protein ,biology.protein ,CRISPR-Cas Systems ,030217 neurology & neurosurgery ,HeLa Cells ,Microsatellite Repeats - Abstract
SummaryTranscription of expanded microsatellite repeats is associated with multiple human diseases, including myotonic dystrophy, Fuchs’ endothelial corneal dystrophy, andC9orf72-ALS/FTD. Eliminating or reducing production of RNA and proteins arising from these expanded loci holds therapeutic benefit. Here, we tested the hypothesis that a deactivated form of the Cas9 enzyme impedes transcription across expanded microsatellites. We observed a repeat length-, PAM-, and strand-dependent reduction in the abundance of repeat-containing RNAs upon targeting dCas9 directly to repeat sequences. Aberrant splicing patterns were rescued in DM1 cells, and production of RAN peptides characteristic of DM1, DM2, andC9orf72-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.
- Published
- 2017
38. An engineered RNA binding protein with improved splicing regulation
- Author
-
Ryan C Day, Melissa Hale, Jared I Richardson, Juan Arboleda, J. Andrew Berglund, Ona L. McConnell, and Eric T. Wang
- Subjects
0301 basic medicine ,RNA-binding protein ,Biology ,Protein Engineering ,03 medical and health sciences ,chemistry.chemical_compound ,Genetics ,RNA Precursors ,RNA and RNA-protein complexes ,MBNL1 ,Humans ,Myotonic Dystrophy ,Nucleotide Motifs ,Zinc finger ,Binding Sites ,Base Sequence ,HEK 293 cells ,Alternative splicing ,RNA ,RNA-Binding Proteins ,Zinc Fingers ,Cell biology ,Alternative Splicing ,030104 developmental biology ,HEK293 Cells ,chemistry ,RNA splicing ,RNA-Binding Motifs ,Binding domain ,HeLa Cells - Abstract
The muscleblind-like (MBNL) family of proteins are key developmental regulators of alternative splicing. Sequestration of MBNL proteins by expanded CUG/CCUG repeat RNA transcripts is a major pathogenic mechanism in the neuromuscular disorder myotonic dystrophy (DM). MBNL1 contains four zinc finger (ZF) motifs that form two tandem RNA binding domains (ZF1–2 and ZF3–4) which each bind YGCY RNA motifs. In an effort to determine the differences in function between these domains, we designed and characterized synthetic MBNL proteins with duplicate ZF1–2 or ZF3–4 domains, referred to as MBNL-AA and MBNL-BB, respectively. Analysis of splicing regulation revealed that MBNL-AA had up to 5-fold increased splicing activity while MBNL-BB had 4-fold decreased activity compared to a MBNL protein with the canonical arrangement of zinc finger domains. RNA binding analysis revealed that the variations in splicing activity are due to differences in RNA binding specificities between the two ZF domains rather than binding affinity. Our findings indicate that ZF1–2 drives splicing regulation via recognition of YGCY RNA motifs while ZF3–4 acts as a general RNA binding domain. Our studies suggest that synthetic MBNL proteins with improved or altered splicing activity have the potential to be used as both tools for investigating splicing regulation and protein therapeutics for DM and other microsatellite diseases.
- Published
- 2017
39. Antisense transcription of the myotonic dystrophy locus yields low-abundant rnas with and without (cag)n repeat
- Author
-
Simon J. van Heeringen, Amanda I de Oude, Bé Wieringa, Derick G. Wansink, Anke E. E. G. Gudde, Joseph Estabrook, Eric T. Wang, and Ingeborg D.G. van Kessel
- Subjects
0301 basic medicine ,Male ,Polyadenylation ,0302 clinical medicine ,Transcription (biology) ,low-abundant RNA ,triplet repeat expansion ,Myotonic Dystrophy ,3' Untranslated Regions ,Antisense RNA ,Genetics ,Myotonin-protein kinase ,bidirectional transcription ,Up-Regulation ,RNA, Long Noncoding ,Transcription Initiation Site ,Molecular Developmental Biology ,Research Paper ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Adolescent ,Other Research Radboud Institute for Molecular Life Sciences [Radboudumc 0] ,Biology ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Cell Line ,03 medical and health sciences ,Open Reading Frames ,RAN translation ,medicine ,Humans ,RNA, Antisense ,RNA, Messenger ,long noncoding RNA ,Molecular Biology ,Alternative splicing ,Intron ,Computational Biology ,Cell Biology ,medicine.disease ,Molecular biology ,Alternative Splicing ,030104 developmental biology ,Case-Control Studies ,microsatellite instability ,Trinucleotide repeat expansion ,Trinucleotide Repeat Expansion ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,030217 neurology & neurosurgery - Abstract
The unstable (CTG·CAG)n trinucleotide repeat in the myotonic dystrophy type 1 (DM1) locus is bidirectionally transcribed from genes with terminal overlap. By transcription in the sense direction, the DMPK gene produces various alternatively spliced mRNAs with a (CUG)n repeat in their 3′ UTR. Expression in opposite orientation reportedly yields (CAG)n-repeat containing RNA, but both structure and biologic significance of this antisense gene (DM1-AS) are largely unknown. Via a combinatorial approach of computational and experimental analyses of RNA from unaffected individuals and DM1 patients we discovered that DM1-AS spans >6 kb, contains alternative transcription start sites and uses alternative polyadenylation sites up- and downstream of the (CAG)n repeat. Moreover, its primary transcripts undergo alternative splicing, whereby the (CAG)n segment is removed as part of an intron. Thus, in patients a mixture of DM1-AS RNAs with and without expanded (CAG)n repeat are produced. DM1-AS expression appears upregulated in patients, but transcript abundance remains very low in all tissues analyzed. Our data suggest that DM1-AS transcripts belong to the class of long non-coding RNAs. These and other biologically relevant implications for how (CAG)n-expanded transcripts may contribute to DM1 pathology can now be explored experimentally.
- Published
- 2017
40. Muscleblind-like 1 (Mbnl1) regulates pre-mRNA alternative splicing during terminal erythropoiesis
- Author
-
Albert W. Cheng, Christopher B. Burge, Paula Trepman, Jiahai Shi, Katherine L. Luo, Eric T. Wang, Piu Wong, Harvey F. Lodish, and Heejo Choi
- Subjects
RNA Splicing Factors ,genetic processes ,Immunology ,Biology ,Biochemistry ,Exon ,Red Cells, Iron, and Erythropoiesis ,hemic and lymphatic diseases ,RNA Precursors ,Animals ,Humans ,natural sciences ,Erythropoiesis ,skin and connective tissue diseases ,Gene knockdown ,Alternative splicing ,RNA-Binding Proteins ,Cell Differentiation ,Cell Biology ,Hematology ,Molecular biology ,Exon skipping ,DNA-Binding Proteins ,Alternative Splicing ,Gene Expression Regulation ,RNA splicing ,sense organs ,Carrier Proteins ,Precursor mRNA - Abstract
The scope and roles of regulated isoform gene expression during erythroid terminal development are poorly understood. We identified hundreds of differentiation-associated isoform changes during terminal erythropoiesis. Sequences surrounding cassette exons of skipped exon events are enriched for motifs bound by the Muscleblind-like (MBNL) family of splicing factors. Knockdown of Mbnl1 in cultured murine fetal liver erythroid progenitors resulted in a strong block in erythroid differentiation and disrupted the developmentally regulated exon skipping of Ndel1 mRNA, which is bound by MBNL1 and critical for erythroid terminal proliferation. These findings reveal an unanticipated scope of the alternative splicing program and the importance of Mbnl1 during erythroid terminal differentiation.
- Published
- 2014
41. Consensus on cerebral involvement in myotonic dystrophy
- Author
-
Carl Morris, Benedikt Schoser, Carmen Alvarez, Enrico Bugiardini, David E. Housman, Bruce M. Wentworth, Louis Richer, John W. Day, Guillaume Bassez, Andrea N. Ladd, Don MacKenzie, Jeffrey R. Wozniak, Nicholas E. Johnson, Gersham Dent, Nathalie Angeard, Christopher E. Pearson, Cynthia Gagnon, Cornelia Kornblum, Nicolas Sergeant, Chad Heatwole, Mat Pletcher, E. Bugiardini, Sita Reddy, Seiji Nishino, Stefan Winblad, G. Meola, Eric T. Wang, Anne Berit Ekström, Laura P.W. Ranum, Geneviè Gourdon, Martina Minnerop, Barbara Fossati, Bruno Eymard, Maurice S. Swanson, and Mário Gomes-Pereira
- Subjects
medicine.medical_specialty ,Pediatrics ,Neurology ,business.industry ,Pediatrics, Perinatology and Child Health ,Physical therapy ,medicine ,Neurology (clinical) ,business ,medicine.disease ,Myotonic dystrophy ,Genetics (clinical) - Published
- 2014
42. Non‐invasive risk assessment of fetal sex chromosome aneuploidy through directed analysis and incorporation of fetal fraction
- Author
-
A. J. Wolfberg, Craig A. Struble, Thomas J. Musci, J. Hooks, Eric T. Wang, K. Juneau, Patrick E. Bogard, Jacob Zahn, Ken Song, M. Mohseni, Arnold Oliphant, and Stephanie Huang
- Subjects
Adult ,medicine.medical_specialty ,Monosomy ,Adolescent ,Aneuploidy ,Biology ,Risk Assessment ,Young Adult ,Fetus ,Sex Factors ,Pregnancy ,Prenatal Diagnosis ,medicine ,Humans ,Genetics (clinical) ,Gynecology ,Sex Chromosomes ,Case-control study ,Obstetrics and Gynecology ,Chromosome ,medicine.disease ,Case-Control Studies ,Cohort ,Female ,False positive rate ,Algorithms - Abstract
Objective To assess the performance of a directed chromosomal analysis approach in the prenatal evaluation of fetal sex chromosome aneuploidy. Methods We analyzed 432 frozen maternal plasma samples obtained from patients prior to undergoing fetal diagnostic testing. The cohort included women greater than 18 years of age with a singleton pregnancy of greater than 10 weeks gestation. Samples were analyzed using a chromosome-selective approach (DANSRTM) and a risk algorithm that incorporates fetal fraction (FORTETM). Results The cohort included 34 cases of sex chromosome aneuploidy. The assay correctly identified 26 of 27 (92.6%) cases of Monosomy X, one case of XXX, and all six cases of XXY. There were four false positive cases of sex chromosome aneuploidy among 380 euploid cases for an overall false positive rate of less than 1%. Discussion Analysis of the risk for sex chromosome aneuploidies can be accomplished with a targeted assay with high sensitivity. © 2014 John Wiley & Sons, Ltd.
- Published
- 2014
43. Contents Vol. 36, 2014
- Author
-
Linda Wu, Andrew McLennan, Jennifer Sanderson, Ahm Kim, Faisal Qureshi, Irene T.M. Lindenburg, Aditi Majajan, Alkan Yildirim, Amanda Henry, Kara Juneau, Linus Bjäreborn, Suzanne M. Jacques, Magnus Nordenskjöld, Alma Aurioles-Garibay, Atıl Yüksel, Inge L. van Kamp, Satz Mengensatzproduktion, Ömer Faruk Demir, N. Scott Adzick, Roberto Romero, Patrick E. Bogard, Karin Pettersson, Mi-Young Lee, Mark P. Johnson, William H. Peranteau, Eric T. Wang, Druckerei Stückle, Gus Ridding, Sonia S. Hassan, Lemi Ibrahimoglu, Ibrahim Kalelioglu, John M.G. van Vugt, Kyu-Sang Kyeong, Harika Yumru, Nahla Khalek, Christopher Kingsley, Craig A. Struble, Annegret Geipel, Jae-Yoon Shim, Christoph Berg, Heiko Reutter, Maynor Garcia, Morassa Mohseni, Aytul Corbacioglu Esmer, Marinus A. Blankenstein, Hayri Ermiş, Tinnakorn Chaiworapongsa, Beverly G. Coleman, Jos W. R. Twisk, Paul Ryvkin, Peter Gustavsson, Lami Yeo, Alan W. Flake, Nikos Papadogiannakis, Cenk Yasa, Neama Meriki, Y. Dogan, Pil Ryang Lee, Recep Has, Ulrich Gembruch, Michael R. Mallmann, Lori J. Howell, Teresa Victoria, Hye-Sung Won, Andreas Müller, Ozlem Dural, Edgar Hernandez-Andrade, Holly L. Hedrick, Jacob Zahn, Jesse D. Vrecenak, Erik Iwarsson, Philip J. Schluter, Jon Hyett, Melanie A. J. Engels, Dick Oepkes, Hyunyoung Ahn, Stephanie Huang, Ellika Sahlin, Arnold Oliphant, Julie S. Moldenhauer, Agne Liedén, Alec W. Welsh, and Thomas M. Boemers
- Subjects
Embryology ,Traditional medicine ,business.industry ,Pediatrics, Perinatology and Child Health ,Obstetrics and Gynecology ,Medicine ,Radiology, Nuclear Medicine and imaging ,General Medicine ,business - Published
- 2014
44. Microarray-Based Cell-Free DNA Analysis Improves Noninvasive Prenatal Testing
- Author
-
Jacob Zahn, Morassa Mohseni, Arnold Oliphant, Paul Ryvkin, Christopher Kingsley, Patrick E. Bogard, Stephanie Huang, Eric T. Wang, Craig A. Struble, and Kara Juneau
- Subjects
Adult ,Embryology ,Down syndrome ,Microarray ,Sequence analysis ,Aneuploidy ,Trisomy ,Prenatal diagnosis ,Computational biology ,Pregnancy ,Prenatal Diagnosis ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Oligonucleotide Array Sequence Analysis ,Genetics ,business.industry ,Obstetrics and Gynecology ,DNA ,Sequence Analysis, DNA ,General Medicine ,medicine.disease ,Cell-free fetal DNA ,Pediatrics, Perinatology and Child Health ,Female ,DNA microarray ,business - Abstract
Objective: To develop a microarray-based method for noninvasive prenatal testing (NIPT) and compare it with next-generation sequencing. Methods: Maternal plasma from 878 pregnant women, including 187 trisomy cases (18 trisomy 13, 37 trisomy 18, 132 trisomy 21), was evaluated for trisomy risk. Targeted chromosomes were analyzed using Digital Analysis of Selected Regions (DANSR™) assays. DANSR products were subsequently divided between two DNA quantification methods: microarrays and next-generation sequencing. For both microarray and sequencing methodologies, the Fetal-Fraction Optimized Risk of Trisomy Evaluation (FORTE™) algorithm was used to determine trisomy risk, assay variability across samples, and compute fetal fraction variability within samples. Results: NIPT using microarrays provided faster and more accurate cell-free DNA (cfDNA) measurements than sequencing. The assay variability, a measure of variance of chromosomal cfDNA counts, was lower for microarrays than for sequencing, 0.051 versus 0.099 (p < 0.0001). Analysis time using microarrays was faster, 7.5 versus 56 h for sequencing. Additionally, fetal fraction precision was improved 1.6-fold by assaying more polymorphic sites with microarrays (p < 0.0001). Microarrays correctly classified all trisomy and nontrisomy cases. Conclusions: NIPT using microarrays delivers more accurate cfDNA analysis than next-generation sequencing and can be performed in less time.
- Published
- 2014
45. A flow cytometry-based screen identifies MBNL1 modulators that rescue splicing defects in myotonic dystrophy type I
- Author
-
Carl Morris, Lauren D. Wood, Mathew T. Pletcher, Lyn H. Jones, Fan Zhang, Eric T. Wang, Nicole E. Bodycombe, Yumei L. Sun, and Keith M. Haskell
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,RNA Splicing ,RNA-binding protein ,Biology ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,Exon ,0302 clinical medicine ,Trinucleotide Repeats ,Genetics ,medicine ,RNA Precursors ,MBNL1 ,Humans ,Myotonic Dystrophy ,Molecular Biology ,3' Untranslated Regions ,Genetics (clinical) ,Three prime untranslated region ,Alternative splicing ,RNA-Binding Proteins ,General Medicine ,Exons ,Articles ,medicine.disease ,Flow Cytometry ,Cell biology ,Alternative Splicing ,030104 developmental biology ,chemistry ,RNA splicing ,Trinucleotide repeat expansion ,Trinucleotide Repeat Expansion ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Myotonic dystrophy Type 1 (DM1) is a rare genetic disease caused by the expansion of CTG trinucleotide repeats ((CTG)exp) in the 3' untranslated region of the DMPK gene. The repeat transcripts sequester the RNA binding protein Muscleblind-like protein 1 (MBNL1) and hamper its normal function in pre-mRNA splicing. Overexpressing exogenous MBNL1 in the DM1 mouse model has been shown to rescue the splicing defects and reverse myotonia. Although a viable therapeutic strategy, pharmacological modulators of MBNL1 expression have not been identified. Here, we engineered a ZsGreen tag into the endogenous MBNL1 locus in HeLa cells and established a flow cytometry-based screening system to identify compounds that increase MBNL1 level. The initial screen of small molecule compound libraries identified more than thirty hits that increased MBNL1 expression greater than double the baseline levels. Further characterization of two hits revealed that the small molecule HDAC inhibitors, ISOX and vorinostat, increased MBNL1 expression in DM1 patient-derived fibroblasts and partially rescued the splicing defect caused by (CUG)exp repeats in these cells. These findings demonstrate the feasibility of this flow-based cytometry screen to identify both small molecule compounds and druggable targets for MBNL1 upregulation.
- Published
- 2016
46. Skeletal Muscle Degenerative Diseases and Strategies for Therapeutic Muscle Repair
- Author
-
Amy J. Wagers, Eric T. Wang, and Mohammadsharif Tabebordbar
- Subjects
medicine.medical_specialty ,Weakness ,Myogenesis ,Skeletal muscle ,Disease ,Biology ,medicine.disease ,Bioinformatics ,Muscular Dystrophies ,Pathology and Forensic Medicine ,Physical medicine and rehabilitation ,medicine.anatomical_structure ,Muscular Diseases ,Sarcopenia ,medicine ,Animals ,Humans ,Muscular dystrophy ,medicine.symptom ,Muscle, Skeletal ,Pathological ,Physiological stress - Abstract
Skeletal muscle is a highly specialized, postmitotic tissue that must withstand chronic mechanical and physiological stress throughout life to maintain proper contractile function. Muscle damage or disease leads to progressive weakness and disability, and manifests in more than 100 different human disorders. Current therapies to treat muscle degenerative diseases are limited mostly to the amelioration of symptoms, although promising new therapeutic directions are emerging. In this review, we discuss the pathological basis for the most common muscle degenerative diseases and highlight new and encouraging experimental and clinical opportunities to prevent or reverse these afflictions.
- Published
- 2013
47. Combinatorial Mutagenesis of MBNL1 Zinc Fingers Elucidates Distinct Classes of Regulatory Events
- Author
-
Jamie Purcell, J. Andrew Berglund, Julia C. Oddo, and Eric T. Wang
- Subjects
Genetics ,RNA Splicing ,Alternative splicing ,Intron ,Exonic splicing enhancer ,RNA-Binding Proteins ,Zinc Fingers ,Prp24 ,Exons ,Articles ,Cell Biology ,Computational biology ,Biology ,Introns ,Exon ,Splicing factor ,SR protein ,Mutagenesis ,RNA splicing ,Humans ,Molecular Biology ,HeLa Cells - Abstract
The RNA binding protein and alternative splicing factor Muscleblind-like 1 (MBNL1) has been a topic of intense study due to its role in myotonic dystrophy (DM) pathogenesis. MBNL1 contains four zinc finger (ZF) RNA binding domains arranged in two pairs. Through combinatorial mutagenesis of the ZF domains, we demonstrate that the pairs of ZFs have differential affinity for RNA and subsequently differential splicing activities. We evaluated splicing and binding activity for six MBNL1-mediated splicing events and found that the splicing activity profiles for the ZF mutants vary among transcripts. Clustering analysis of splicing profiles revealed that two distinct classes of MBNL1 pre-mRNA substrates exist. For some of the RNA transcripts tested, binding and splicing activity of the ZF mutants correlated. However, for some transcripts it appears that MBNL1 exerts robust splicing activity in the absence of RNA binding. We demonstrate that functionally distinct classes of MBNL1-mediated splicing events exist as defined by requirements for ZF-RNA interactions.
- Published
- 2012
48. Design of a Bioactive Small Molecule That Targets the Myotonic Dystrophy Type 1 RNA via an RNA Motif–Ligand Database and Chemical Similarity Searching
- Author
-
Amit Kumar, David E. Housman, Raman Parkesh, Thomas D. Wang, Eric T. Wang, Jessica L. Childs-Disney, Jason W. Hoskins, Matthew D. Disney, Charles A. Thornton, Masayuki Nakamori, and Tuan Tran
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Databases, Factual ,RNA-binding protein ,Ligands ,Biochemistry ,Myotonic dystrophy ,Article ,Catalysis ,Small Molecule Libraries ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,medicine ,Humans ,Myotonic Dystrophy ,MBNL1 ,Gene ,biology ,Chemistry ,RNA-Binding Proteins ,RNA ,General Chemistry ,medicine.disease ,Small molecule ,Insulin receptor ,RNA splicing ,biology.protein ,HeLa Cells - Abstract
Myotonic dystrophy type 1 (DM1) is a triplet repeating disorder caused by expanded CTG repeats in the 3'-untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. The transcribed repeats fold into an RNA hairpin with multiple copies of a 5'CUG/3'GUC motif that binds the RNA splicing regulator muscleblind-like 1 protein (MBNL1). Sequestration of MBNL1 by expanded r(CUG) repeats causes splicing defects in a subset of pre-mRNAs including the insulin receptor, the muscle-specific chloride ion channel, sarco(endo)plasmic reticulum Ca(2+) ATPase 1, and cardiac troponin T. Based on these observations, the development of small-molecule ligands that target specifically expanded DM1 repeats could be of use as therapeutics. In the present study, chemical similarity searching was employed to improve the efficacy of pentamidine and Hoechst 33258 ligands that have been shown previously to target the DM1 triplet repeat. A series of in vitro inhibitors of the RNA-protein complex were identified with low micromolar IC(50)'s, which are20-fold more potent than the query compounds. Importantly, a bis-benzimidazole identified from the Hoechst query improves DM1-associated pre-mRNA splicing defects in cell and mouse models of DM1 (when dosed with 1 mM and 100 mg/kg, respectively). Since Hoechst 33258 was identified as a DM1 binder through analysis of an RNA motif-ligand database, these studies suggest that lead ligands targeting RNA with improved biological activity can be identified by using a synergistic approach that combines analysis of known RNA-ligand interactions with chemical similarity searching.
- Published
- 2012
49. Selective analysis of cell-free DNA in maternal blood for evaluation of fetal trisomy
- Author
-
Naiping Shen, Michel Sun, Andrew B. Sparks, Ken Song, Jill Garrison, Patrick Pattee, Aoy Tomita-Mitchell, Craig A. Struble, Jacob Zahn, Jay Sandler, Celeste McBride, Renee Stokowski, John R. Stuelpnagel, Desiree Hollemon, Kevin J. Lee, Michael S Mitchell, Arnold Oliphant, Eric T. Wang, Wade A. Barrett, and Jigna Doshi
- Subjects
medicine.diagnostic_test ,Shotgun sequencing ,Obstetrics and Gynecology ,Aneuploidy ,Prenatal diagnosis ,Biology ,medicine.disease ,Molecular biology ,DNA sequencing ,Deep sequencing ,Cell-free fetal DNA ,medicine ,Trisomy ,Genetics (clinical) ,Genetic testing - Abstract
Objective To develop a novel prenatal assay based on selective analysis of cell-free DNA in maternal blood for evaluation of fetal Trisomy 21 (T21) and Trisomy 18 (T18). Methods Two hundred ninety-eight pregnancies, including 39 T21 and seven T18 confirmed fetal aneuploidies, were analyzed using a novel, highly multiplexed assay, termed digital analysis of selected regions (DANSR ™ ). Cell-free DNA from maternal blood samples was analyzed using DANSR assays for loci on chromosomes 21 and 18. Products from 96 separate patients were pooled and sequenced together. A standard Z-test of chromosomal proportions was used to distinguish aneuploid samples from average-risk pregnancy samples. DANSR aneuploidy discrimination was evaluated at various sequence depths. Results At the lowest sequencing depth, corresponding to 204000 sequencing counts per sample, average-risk cases where distinguished from T21 and T18 cases, with Z statistics for all cases exceeding 3.6. Increasing the sequencing depth to 410000 counts per sample substantially improved separation of aneuploid and average-risk cases. A further increase to 620000 counts per sample resulted in only marginal improvement. This depth of sequencing represents less than 5% of that required by massively parallel shotgun sequencing approaches. Conclusion Digital analysis of selected regions enables highly accurate, cost efficient, and scalable noninvasive fetal aneuploidy assessment. © 2012 John Wiley & Sons, Ltd.
- Published
- 2012
50. Triple Degradation of BTK, IKZF1 and IKZF3 in B-Cell Malignancies
- Author
-
Nathanael S. Gray, Sara N. Morrow, Eric T. Wang, Guang Yang, Dennis Dobrovolsky, Eric S. Fischer, Katherine A. Donovan, Zhengnian Li, Tyler B. Faust, Radosław P. Nowak, Steven P. Treon, and David M. Weinstock
- Subjects
biology ,Kinase ,Bortezomib ,Chronic lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,Protein degradation ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,chemistry ,immune system diseases ,hemic and lymphatic diseases ,Ibrutinib ,Cancer research ,biology.protein ,medicine ,Bruton's tyrosine kinase ,Mantle cell lymphoma ,Tyrosine kinase ,medicine.drug - Abstract
Bruton's Tyrosine Kinase (BTK), a TEC-family non-receptor tyrosine kinase, plays a critical role in B-cell development and function. Targeting of BTK with covalent inhibitors like ibrutinib has become a standard approach for many B-cell malignancies, including chronic lymphocytic leukemia (CLL), Waldenstrom macroglobulinemia, mantle cell lymphoma (MCL) and marginal zone lymphoma (MZL). Yet, many patients demonstrate intrinsic or acquired resistance to covalent BTK inhibition. The prognosis of patients who relapse after ibrutinib treatment for MCL or CLL is dismal, highlighting the urgent need for new approaches that overcome resistance to current BTK inhibitors. We hypothesized that degradation of BTK could be a better alternative to inhibition alone, as it would both: 1) maintain efficacy in cells harboring the ibrutinib-resistant BTK C418S mutation and 2) target non-catalytic functions of BTK. To address this, we turned to a small molecule-mediated protein degradation platform that utilizes an E3 ligase-targeting moiety linked to the ligand of a target of interest, so that the target can be marked for ubiquitination and subsequent proteasomal degradation. We previously showed that BTK is one of the most robustly degraded kinase targets using a nonspecific kinase inhibitor linked to a imide-based core followed by agnostic proteomics. We synthesized highly potent and selective degraders of BTK using imides as a base. To do so, the parent BTK inhibitor CGI1746 was linked to thalidomide using either polyethylene glycol (DD-03-007) linkers or saturated hydrocarbon chain (DD-03-171) linkers. After verifying that these degraders induce dimerization of BTK and CRBN and penetrate cells, we explored the pharmacological effects in vitro. Both DD-03-007 and DD-03-171 reduced BTK levels at concentrations as low as 40 nM within 4h of treatment. Furthermore, cellular BTK levels remained low for 24h after washout, showing that these degraders are capable of sustaining depletion of BTK for an extended period of time (Figure). DD-03-007 and DD-03-171 are both ligase and proteasome-dependent, as shown by co-treatment with bortezomib or MLN-4924 as well as competition experiments with lenalidomide or CGI-1746. Moreover, the degraders exhibited strong synergy with the HCK inhibitor A419259, suggesting that it would be possible to recapitulate ibrutinib's previously reported polypharmacology with an HCK inhibitor. We overexpressed wild-type or C481S BTK in TMD-8 cells and ran an antiproliferation assay, which showed that DD-03-007 overcame ibrutinib resistance associated with BTK C481S mutation. Next, we explored the effects of the degraders in MCL specifically. Proteomic analysis showed that DD-03-171 is a triple-degrader, as it degrades BTK but retains degradation activity on IKZF1 and IKZF3. Finally, we performed In vivo efficacy studies in cell line and patient-derived xenograft (PDX) models of MCL. The latter was obtained from a patient who had progressed on ibrutinib. In both models, DD-03-171 caused a significant reduction in tumor burden at an early timepoint. DD-03-171 also markedly extended survival compared to treatment with ibrutinib or lenalidomide alone (Figure). In conclusion, we developed highly potent and selective BTK degraders with activity in vivo against human MCL that induce the degradation of multiple factors essential for MCL survival. Figure. Figure. Disclosures Treon: Pharmacyclics: Consultancy, Other: Travel, Accommodations, Expenses, Research Funding; Janssen: Consultancy, Other: Travel, Accommodations, Expenses; Johnson & Johnson: Consultancy; BMS: Research Funding. Weinstock:Novartis, Dragonfly, Travera, DxTerity, Travera: Consultancy; Novartis: Consultancy, Research Funding; Novartis, Astra Zeneca, Abbvie, Aileron, Surface Oncology, Daiichi Sankyo: Research Funding; Genentech/Roche, Monsanto: Consultancy; Astra Zeneca, JAX, Samumed, Regeneron, Sun Pharma, Prescient: Patents & Royalties; Travera: Equity Ownership. Gray:Syros, Soltego, Petra, C4 Therapeutics: Equity Ownership.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.