1
|
Abati E, Alberti C, Tambè V, Esseridou A, Comi GP, Corti S, Meola G, Secchi F. Cardiac risk and myocardial fibrosis assessment with cardiac magnetic resonance in patients with myotonic dystrophy. Front Neurol 2024; 15:1493570. [PMID: 39639989 PMCID: PMC11617365 DOI: 10.3389/fneur.2024.1493570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Non-invasive evaluation of myocardial tissue is a major goal of cardiac imaging. This is the case of myocardial fibrosis which is crucial in many myocardial diseases. Cardiac extracellular volume (ECV) was shown to indicate myocardial fibrosis and early cardiac involvement. With this study, our objective is to evaluate ECV measured with cardiac magnetic resonance (CMR) in patients with myotonic dystrophy type 1 (DM1) and 2 (DM2) as potential imaging biomarkers of subclinical cardiac pathology, and its relationship with demographic and clinical parameters, ECG-derived measures of cardiac conduction, and neuromuscular performance status. Materials and methods We retrospectively analyzed 18 DM1 patients and 4 DM2 patients without apparent cardiac disease who had CMR at our center. Differences between independent distributions were evaluated using Mann-Whitney U test, while correlations were evaluated using Spearman's ρ. Results Global ECV in DM1 patients (median 28.36; IQR 24.81-29.77) was significantly higher (p = 0.0141) than in DM2 patients (median 22.93; IQR 21.25-24.35), and than that reported in literature in healthy subjects (p = 0.0374; median 25.60; IQR 19.90-31.90). Septal ECV was significantly higher (p = 0.0074) in DM1 (median 27.37; IQR 25.97-29.74) than in DM2 patients (median 22.46; 21.57-23.19). Global ECV showed a strong, positive correlation with septal ECV (ρ = 0.9282, p < 0.0001). We observed that DM1 women showed significantly higher global (p = 0.0012) and septal (p < 0.0001) ECV values compared to men. Discussion We found a significant increase in global and septal cardiac ECV in patients with DM1. These values might thus suggest that DM1 patients present an increased cardiovascular risk, mainly due to cardiac fibrosis, even in absence of overt cardiac pathology at other common cardiovascular exams. DM1 patients may also be at increased risk of early septal fibrosis, with important implications on the risk for fatal arrhythmias. In addition, our results suggest the presence of gender-related differences, with DM1 women being more prone to myocardial fibrosis. Physicians dealing with DM1 may consider CMR as a screening tool for the early identification of patients with increased cardiovascular risk.
Collapse
Affiliation(s)
- Elena Abati
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudia Alberti
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | | | | | - Giacomo Pietro Comi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
- Neurology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Meola
- Department of Neurorehabilitation Sciences, Casa Di Cura Igea, Milan, Italy
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Francesco Secchi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
- Unit of Cardiovascular Imaging, IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
2
|
Petri H, Mohammad BJY, Kristensen AT, Thune JJ, Vissing J, Køber L, Witting N, Bundgaard H, Christensen AH. Natural history of cardiac involvement in myotonic dystrophy type 1 - Emphasis on the need for lifelong follow-up. Int J Cardiol 2024; 406:132070. [PMID: 38643802 DOI: 10.1016/j.ijcard.2024.132070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/19/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Cardiac involvement represents a major cause of morbidity and mortality in patients with myotonic dystrophy type 1 (DM1) and prevention of sudden cardiac death (SCD) is a central part of patient care. We investigated the natural history of cardiac involvement in patients with DM1 to provide an evidence-based foundation for adjustment of follow-up protocols. METHODS Patients with genetically confirmed DM1 were identified. Data on patient characteristics, performed investigations (12 lead ECG, Holter monitoring and echocardiography), and clinical outcomes were retrospectively collected from electronic health records. RESULTS We included 195 patients (52% men) with a mean age at baseline evaluation of 41 years (range 14-79). The overall prevalence of cardiac involvement increased from 42% to 66% after a median follow-up of 10.5 years. There was a male predominance for cardiac involvement at end of follow-up (74 vs. 44%, p < 0.001). The most common types of cardiac involvement were conduction abnormalities (48%), arrhythmias (35%), and left ventricular systolic dysfunction (21%). Only 17% of patients reported cardiac symptoms. The standard 12‑lead ECG was the most sensitive diagnostic modality and documented cardiac involvement in 24% at baseline and in 49% at latest follow-up. However, addition of Holter monitoring and echocardiography significantly increased the diagnostic yield with 18 and 13% points at baseline and latest follow-up, respectively. Despite surveillance 35 patients (18%) died during follow-up; seven due to SCD. CONCLUSIONS In patients with DM1 cardiac involvement was highly prevalent and developed during follow-up. These findings justify lifelong follow-up with ECG, Holter, and echocardiography. CLINICAL PERSPECTIVE What is new? What are the clinical implications?
Collapse
Affiliation(s)
- Helle Petri
- Department of Cardiology, Copenhagen University Hospital - Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Batool J Y Mohammad
- Department of Cardiology, Copenhagen University Hospital - Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Andreas Torp Kristensen
- Department of Cardiology, Copenhagen University Hospital - Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Jens Jakob Thune
- Department of Cardiology, Copenhagen University Hospital - Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Lars Køber
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Nanna Witting
- Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Alex Hørby Christensen
- Department of Cardiology, Copenhagen University Hospital - Herlev-Gentofte Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
3
|
Angom RS, Joshi A, Patowary A, Sivadas A, Ramasamy S, K. V. S, Kaushik K, Sabharwal A, Lalwani MK, K. S, Singh N, Scaria V, Sivasubbu S. Forward genetic screen using a gene-breaking trap approach identifies a novel role of grin2bb-associated RNA transcript ( grin2bbART) in zebrafish heart function. Front Cell Dev Biol 2024; 12:1339292. [PMID: 38533084 PMCID: PMC10964321 DOI: 10.3389/fcell.2024.1339292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
LncRNA-based control affects cardiac pathophysiologies like myocardial infarction, coronary artery disease, hypertrophy, and myotonic muscular dystrophy. This study used a gene-break transposon (GBT) to screen zebrafish (Danio rerio) for insertional mutagenesis. We identified three insertional mutants where the GBT captured a cardiac gene. One of the adult viable GBT mutants had bradycardia (heart arrhythmia) and enlarged cardiac chambers or hypertrophy; we named it "bigheart." Bigheart mutant insertion maps to grin2bb or N-methyl D-aspartate receptor (NMDAR2B) gene intron 2 in reverse orientation. Rapid amplification of adjacent cDNA ends analysis suggested a new insertion site transcript in the intron 2 of grin2bb. Analysis of the RNA sequencing of wild-type zebrafish heart chambers revealed a possible new transcript at the insertion site. As this putative lncRNA transcript satisfies the canonical signatures, we called this transcript grin2bb associated RNA transcript (grin2bbART). Using in situ hybridization, we confirmed localized grin2bbART expression in the heart, central nervous system, and muscles in the developing embryos and wild-type adult zebrafish atrium and bulbus arteriosus. The bigheart mutant had reduced Grin2bbART expression. We showed that bigheart gene trap insertion excision reversed cardiac-specific arrhythmia and atrial hypertrophy and restored grin2bbART expression. Morpholino-mediated antisense downregulation of grin2bbART in wild-type zebrafish embryos mimicked bigheart mutants; this suggests grin2bbART is linked to bigheart. Cardiovascular tissues use Grin2bb as a calcium-permeable ion channel. Calcium imaging experiments performed on bigheart mutants indicated calcium mishandling in the heart. The bigheart cardiac transcriptome showed differential expression of calcium homeostasis, cardiac remodeling, and contraction genes. Western blot analysis highlighted Camk2d1 and Hdac1 overexpression. We propose that altered calcium activity due to disruption of grin2bbART, a putative lncRNA in bigheart, altered the Camk2d-Hdac pathway, causing heart arrhythmia and hypertrophy in zebrafish.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, United States
| | - Adita Joshi
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Ashok Patowary
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Ambily Sivadas
- GN Ramachandran Knowledge Center for Genome Informatics, Council of Scientific and Industrial Research, Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Soundhar Ramasamy
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Shamsudheen K. V.
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- GN Ramachandran Knowledge Center for Genome Informatics, Council of Scientific and Industrial Research, Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Kriti Kaushik
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Ankit Sabharwal
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Mukesh Kumar Lalwani
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Subburaj K.
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Naresh Singh
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Vinod Scaria
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- GN Ramachandran Knowledge Center for Genome Informatics, Council of Scientific and Industrial Research, Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sridhar Sivasubbu
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
4
|
Yadava RS, Mandal M, Mahadevan MS. Studying the Effect of MBNL1 and MBNL2 Loss in Skeletal Muscle Regeneration. Int J Mol Sci 2024; 25:2687. [PMID: 38473933 PMCID: PMC10931579 DOI: 10.3390/ijms25052687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Loss of function of members of the muscleblind-like (MBNL) family of RNA binding proteins has been shown to play a key role in the spliceopathy of RNA toxicity in myotonic dystrophy type 1 (DM1), the most common muscular dystrophy affecting adults and children. MBNL1 and MBNL2 are the most abundantly expressed members in skeletal muscle. A key aspect of DM1 is poor muscle regeneration and repair, leading to dystrophy. We used a BaCl2-induced damage model of muscle injury to study regeneration and effects on skeletal muscle satellite cells (MuSCs) in Mbnl1∆E3/∆E3 and Mbnl2∆E2/∆E2 knockout mice. Similar experiments have previously shown deleterious effects on these parameters in mouse models of RNA toxicity. Muscle regeneration in Mbnl1 and Mbnl2 knockout mice progressed normally with no obvious deleterious effects on MuSC numbers or increased expression of markers of fibrosis. Skeletal muscles in Mbnl1∆E3/∆E3/ Mbnl2∆E2/+ mice showed increased histopathology but no deleterious reductions in MuSC numbers and only a slight increase in collagen deposition. These results suggest that factors beyond the loss of MBNL1/MBNL2 and the associated spliceopathy are likely to play a key role in the defects in skeletal muscle regeneration and deleterious effects on MuSCs that are seen in mouse models of RNA toxicity due to expanded CUG repeats.
Collapse
Affiliation(s)
| | | | - Mani S. Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA; (R.S.Y.)
| |
Collapse
|
5
|
Luo L, Li Y, Bao Z, Zhu D, Chen G, Li W, Xiao Y, Wang Z, Zhang Y, Liu H, Chen Y, Liao Y, Cheng K, Li Z. Pericardial Delivery of SDF-1α Puerarin Hydrogel Promotes Heart Repair and Electrical Coupling. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2302686. [PMID: 37665792 DOI: 10.1002/adma.202302686] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/02/2023] [Indexed: 09/06/2023]
Abstract
The stromal-derived factor 1α/chemokine receptor 4 (SDF-1α/CXCR4) axis contributes to myocardial protection after myocardial infarction (MI) by recruiting endogenous stem cells into the ischemic tissue. However, excessive inflammatory macrophages are also recruited simultaneously, aggravating myocardial damage. More seriously, the increased inflammation contributes to abnormal cardiomyocyte electrical coupling, leading to inhomogeneities in ventricular conduction and retarded conduction velocity. It is highly desirable to selectively recruit the stem cells but block the inflammation. In this work, SDF-1α-encapsulated Puerarin (PUE) hydrogel (SDF-1α@PUE) is capable of enhancing endogenous stem cell homing and simultaneously polarizing the recruited monocyte/macrophages into a repairing phenotype. Flow cytometry analysis of the treated heart tissue shows that endogenous bone marrow mesenchymal stem cells, hemopoietic stem cells, and immune cells are recruited while SDF-1α@PUE efficiently polarizes the recruited monocytes/macrophages into the M2 type. These macrophages influence the preservation of connexin 43 (Cx43) expression which modulates intercellular coupling and improves electrical conduction. Furthermore, by taking advantage of the improved "soil", the recruited stem cells mediate an improved cardiac function by preventing deterioration, promoting neovascular architecture, and reducing infarct size. These findings demonstrate a promising therapeutic platform for MI that not only facilitates heart regeneration but also reduces the risk of cardiac arrhythmias.
Collapse
Affiliation(s)
- Li Luo
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| | - Yuetong Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Ziwei Bao
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27606, USA
| | - Guoqin Chen
- Cardiology Department of Panyu Central Hospital and Cardiovascular Disease Institute of Panyu District, Guangzhou, 511400, P. R. China
| | - Weirun Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| | - Yingxian Xiao
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| | - Zhenzhen Wang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27606, USA
| | - Yixin Zhang
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding, 071002, China
| | - Yanmei Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, 10032, USA
| | - Zhenhua Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510515, China
| |
Collapse
|
6
|
Zhu X, Hao J, Zhang H, Chi M, Wang Y, Huang J, Xu R, Xincai Z, Xin B, Sun X, Zhang J, Zhou S, Cheng D, Yuan T, Ding J, Zheng S, Guo C, Yang Q. Oncometabolite D-2-hydroxyglutarate-dependent metabolic reprogramming induces skeletal muscle atrophy during cancer cachexia. Commun Biol 2023; 6:977. [PMID: 37741882 PMCID: PMC10518016 DOI: 10.1038/s42003-023-05366-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
Cancer cachexia is characterized by weight loss and skeletal muscle wasting. Based on the up-regulation of catabolism and down-regulation of anabolism, here we showed genetic mutation-mediated metabolic reprogramming in the progression of cancer cachexia by screening for metabolites and investigating their direct effect on muscle atrophy. Treatment with 93 μM D-2-hydroxyglutarate (D2HG) resulted in reduced myotube width and increased expression of E3 ubiquitin ligases. Isocitrate Dehydrogenase 1 (IDH1) mutant patients had higher D2HG than non-mutant patients. In the in vivo murine cancer cachexia model, mutant IDH1 in CT26 cancer cells accelerated cachexia progression and worsened overall survival. Transcriptomics and metabolomics revealed a distinct D2HG-induced metabolic imbalance. Treatment with the IDH1 inhibitor ivosidenib delayed the progression of cancer cachexia in murine GL261 glioma model and CT26 colorectal carcinoma models. These data demonstrate the contribution of IDH1 mutation mediated D2HG accumulation to the progression of cancer cachexia and highlight the individualized treatment of IDH1 mutation associated cancer cachexia.
Collapse
Affiliation(s)
- Xinting Zhu
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Juan Hao
- Department of Endocrinology, Shanghai Traditional Chinese Medicine, Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 230 Baoding Road, Shanghai, 200082, China
| | - Hong Zhang
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Mengyi Chi
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yaxian Wang
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Jinlu Huang
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Rong Xu
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zhao Xincai
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Bo Xin
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Xipeng Sun
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Jianping Zhang
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Shumin Zhou
- Institution of microsurgery on extremities, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Dongdong Cheng
- Department of Bone Oncology, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of MedicineShanghai Shanghai, Shanghai, P. R. China
| | - Ting Yuan
- Department of Bone Oncology, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of MedicineShanghai Shanghai, Shanghai, P. R. China
| | - Jun Ding
- Department of Neurosurgery, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Shuier Zheng
- Department of Oncology, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Cheng Guo
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Quanjun Yang
- Department of Pharmacy, Shanghai Sixth People's Hospital affiliated Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
7
|
Pierre M, Djemai M, Chapotte-Baldacci CA, Pouliot V, Puymirat J, Boutjdir M, Chahine M. Cardiac involvement in patient-specific induced pluripotent stem cells of myotonic dystrophy type 1: unveiling the impact of voltage-gated sodium channels. Front Physiol 2023; 14:1258318. [PMID: 37791351 PMCID: PMC10544896 DOI: 10.3389/fphys.2023.1258318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a genetic disorder that causes muscle weakness and myotonia. In DM1 patients, cardiac electrical manifestations include conduction defects and atrial fibrillation. DM1 results in the expansion of a CTG transcribed into CUG-containing transcripts that accumulate in the nucleus as RNA foci and alter the activity of several splicing regulators. The underlying pathological mechanism involves two key RNA-binding proteins (MBNL and CELF) with expanded CUG repeats that sequester MBNL and alter the activity of CELF resulting in spliceopathy and abnormal electrical activity. In the present study, we identified two DM1 patients with heart conduction abnormalities and characterized their hiPSC lines. Two differentiation protocols were used to investigate both the ventricular and the atrial electrophysiological aspects of DM1 and unveil the impact of the mutation on voltage-gated ion channels, electrical activity, and calcium homeostasis in DM1 cardiomyocytes derived from hiPSCs. Our analysis revealed the presence of molecular hallmarks of DM1, including the accumulation of RNA foci and sequestration of MBNL1 in DM1 hiPSC-CMs. We also observed mis-splicing of SCN5A and haploinsufficiency of DMPK. Furthermore, we conducted separate characterizations of atrial and ventricular electrical activity, conduction properties, and calcium homeostasis. Both DM1 cell lines exhibited reduced density of sodium and calcium currents, prolonged action potential duration, slower conduction velocity, and impaired calcium transient propagation in both ventricular and atrial cardiomyocytes. Notably, arrhythmogenic events were recorded, including both ventricular and atrial arrhythmias were observed in the two DM1 cell lines. These findings enhance our comprehension of the molecular mechanisms underlying DM1 and provide valuable insights into the pathophysiology of ventricular and atrial involvement.
Collapse
Affiliation(s)
| | | | | | | | - Jack Puymirat
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, QC, Canada
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
- Departments of Cell Biology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Mohamed Chahine
- CERVO Research Center, Quebec City, QC, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
8
|
Porta-Sánchez A, Priori SG. Genetic Abnormalities of the Sinoatrial Node and Atrioventricular Conduction. Cardiol Clin 2023; 41:333-347. [PMID: 37321685 DOI: 10.1016/j.ccl.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The peculiar electrophysiological properties of the sinoatrial node and the cardiac conduction system are key components of the normal physiology of cardiac impulse generation and propagation. Multiple genes and transcription factors and metabolic proteins are involved in their development and regulation. In this review, we have summarized the genetic underlying causes, key clinical findings, and the latest available clinical evidence. We will discuss clinical diagnosis and management of the genetic conditions associated with conduction disorders that are more prevalent in clinical practice, for this reason, very rare genetic diseases presenting sinus node or cardiac conduction system abnormalities are not discussed.
Collapse
Affiliation(s)
- Andreu Porta-Sánchez
- Cardiología Molecular, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid, Spain; Departamento de Cardiología, Unidad de Arritmias, Hospital Universitario Quironsalud Madrid, Spain; Departamento de Medicina, Universidad Europea de Madrid, Spain
| | - Silvia Giuliana Priori
- Cardiología Molecular, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid, Spain; Molecular Medicine Department, University of Pavia, Italy; Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy.
| |
Collapse
|
9
|
McBride D, Deshmukh A, Shore S, Elafros MA, Liang JJ. Cardiac Involvement and Arrhythmias Associated with Myotonic Dystrophy. Rev Cardiovasc Med 2022; 23:126. [PMID: 36177340 PMCID: PMC9518819 DOI: 10.31083/j.rcm2304126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/28/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Myotonic dystrophy is an autosomal dominant genetic disease of nucleotide expansion resulting in neuromuscular disease with two distinct subtypes. There are significant systemic manifestations of this condition including progressive muscular decline, neurologic abnormalities, and cardiac disease. Given the higher prevalence of cardiac dysfunction compared to the general population, there is significant interest in early diagnosis and prevention of cardiac morbidity and mortality. Cardiac dysfunction has an origin in abnormal and unstable nucleotide repeats in the DMPK and CNBP genes which have downstream effects leading to an increased propensity for arrhythmias and left ventricular systolic dysfunction. Current screening paradigms involve the use of routine screening electrocardiograms, ambulatory electrocardiographic monitors, and cardiac imaging to stratify risk and suggest further invasive evaluation. The most common cardiac abnormality is atrial arrhythmia, however there is significant mortality in this population from high-degree atrioventricular block and ventricular arrhythmia. In this review, we describe the cardiac manifestations of myotonic dystrophy with an emphasis on arrhythmia which is the second most common cause of death in this population after respiratory failure.
Collapse
Affiliation(s)
- Daniel McBride
- Electrophysiology Section, Division of Cardiology, Ann Arbor, MI 48109, USA
| | - Amrish Deshmukh
- Electrophysiology Section, Division of Cardiology, Ann Arbor, MI 48109, USA
| | - Supriya Shore
- Heart Failure Section, Division of Cardiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Melissa A. Elafros
- Neuromuscular Section, Division of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jackson J. Liang
- Electrophysiology Section, Division of Cardiology, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Rocha CT, Escolar DM. Treatment and Management of Muscular Dystrophies. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Porta-Sánchez A, Priori SG. Genetic Abnormalities of the Sinoatrial Node and Atrioventricular Conduction. Card Electrophysiol Clin 2021; 13:625-639. [PMID: 34689891 DOI: 10.1016/j.ccep.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The peculiar electrophysiological properties of the sinoatrial node and the cardiac conduction system are key components of the normal physiology of cardiac impulse generation and propagation. Multiple genes and transcription factors and metabolic proteins are involved in their development and regulation. In this review, we have summarized the genetic underlying causes, key clinical findings, and the latest available clinical evidence. We will discuss clinical diagnosis and management of the genetic conditions associated with conduction disorders that are more prevalent in clinical practice, for this reason, very rare genetic diseases presenting sinus node or cardiac conduction system abnormalities are not discussed.
Collapse
Affiliation(s)
- Andreu Porta-Sánchez
- Cardiología Molecular, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid, Spain; Departamento de Cardiología, Unidad de Arritmias, Hospital Universitario Quironsalud Madrid, Spain; Departamento de Medicina, Universidad Europea de Madrid, Spain
| | - Silvia Giuliana Priori
- Cardiología Molecular, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) Madrid, Spain; Molecular Medicine Department, University of Pavia, Italy; Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy.
| |
Collapse
|
12
|
Etheridge SP, Niu MC. Electrophysiologic Consequences and Sudden Death in Myotonic Dystrophy: Beyond the ECG and Toward Precision Medicine. JACC Clin Electrophysiol 2021; 7:1049-1051. [PMID: 34412868 DOI: 10.1016/j.jacep.2021.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Susan P Etheridge
- Department of Pediatrics, University of Utah and Primary Children's Hospital, Salt Lake City, Utah, USA.
| | - Mary C Niu
- Department of Pediatrics, University of Utah and Primary Children's Hospital, Salt Lake City, Utah, USA
| |
Collapse
|
13
|
Ozimski LL, Sabater-Arcis M, Bargiela A, Artero R. The hallmarks of myotonic dystrophy type 1 muscle dysfunction. Biol Rev Camb Philos Soc 2020; 96:716-730. [PMID: 33269537 DOI: 10.1111/brv.12674] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is the most prevalent form of muscular dystrophy in adults and yet there are currently no treatment options. Although this disease causes multisystemic symptoms, it is mainly characterised by myopathy or diseased muscles, which includes muscle weakness, atrophy, and myotonia, severely affecting the lives of patients worldwide. On a molecular level, DM1 is caused by an expansion of CTG repeats in the 3' untranslated region (3'UTR) of the DM1 Protein Kinase (DMPK) gene which become pathogenic when transcribed into RNA forming ribonuclear foci comprised of auto complementary CUG hairpin structures that can bind proteins. This leads to the sequestration of the muscleblind-like (MBNL) family of proteins, depleting them, and the abnormal stabilisation of CUGBP Elav-like family member 1 (CELF1), enhancing it. Traditionally, DM1 research has focused on this RNA toxicity and how it alters MBNL and CELF1 functions as key splicing regulators. However, other proteins are affected by the toxic DMPK RNA and there is strong evidence that supports various signalling cascades playing an important role in DM1 pathogenesis. Specifically, the impairment of protein kinase B (AKT) signalling in DM1 increases autophagy, apoptosis, and ubiquitin-proteasome activity, which may also be affected in DM1 by AMP-activated protein kinase (AMPK) downregulation. AKT also regulates CELF1 directly, by affecting its subcellular localisation, and indirectly as it inhibits glycogen synthase kinase 3 beta (GSK3β), which stabilises the repressive form of CELF1 in DM1. Another kinase that contributes to CELF1 mis-regulation, in this case by hyperphosphorylation, is protein kinase C (PKC). Additionally, it has been demonstrated that fibroblast growth factor-inducible 14 (Fn14) is induced in DM1 and is associated with downstream signalling through the nuclear factor κB (NFκB) pathways, associating inflammation with this disease. Furthermore, MBNL1 and CELF1 play a role in cytoplasmic processes involved in DM1 myopathy, altering proteostasis and sarcomere structure. Finally, there are many other elements that could contribute to the muscular phenotype in DM1 such as alterations to satellite cells, non-coding RNA metabolism, calcium dysregulation, and repeat-associated non-ATG (RAN) translation. This review aims to organise the currently dispersed knowledge on the different pathways affected in DM1 and discusses the unexplored connections that could potentially help in providing new therapeutic targets in DM1 research.
Collapse
Affiliation(s)
- Lauren L Ozimski
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain.,Arthex Biotech, Catedrático Escardino, 9, Paterna, Valencia, 46980, Spain
| | - Maria Sabater-Arcis
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain
| | - Ruben Artero
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain
| |
Collapse
|
14
|
Sztretye M, Szabó L, Dobrosi N, Fodor J, Szentesi P, Almássy J, Magyar ZÉ, Dienes B, Csernoch L. From Mice to Humans: An Overview of the Potentials and Limitations of Current Transgenic Mouse Models of Major Muscular Dystrophies and Congenital Myopathies. Int J Mol Sci 2020; 21:ijms21238935. [PMID: 33255644 PMCID: PMC7728138 DOI: 10.3390/ijms21238935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Muscular dystrophies are a group of more than 160 different human neuromuscular disorders characterized by a progressive deterioration of muscle mass and strength. The causes, symptoms, age of onset, severity, and progression vary depending on the exact time point of diagnosis and the entity. Congenital myopathies are rare muscle diseases mostly present at birth that result from genetic defects. There are no known cures for congenital myopathies; however, recent advances in gene therapy are promising tools in providing treatment. This review gives an overview of the mouse models used to investigate the most common muscular dystrophies and congenital myopathies with emphasis on their potentials and limitations in respect to human applications.
Collapse
|
15
|
Yadava RS, Yu Q, Mandal M, Rigo F, Bennett CF, Mahadevan MS. Systemic therapy in an RNA toxicity mouse model with an antisense oligonucleotide therapy targeting a non-CUG sequence within the DMPK 3'UTR RNA. Hum Mol Genet 2020; 29:1440-1453. [PMID: 32242217 PMCID: PMC7268549 DOI: 10.1093/hmg/ddaa060] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/19/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most common adult muscular dystrophy, is an autosomal dominant disorder caused by an expansion of a (CTG)n tract within the 3' untranslated region (3'UTR) of the dystrophia myotonica protein kinase (DMPK) gene. Mutant DMPK mRNAs are toxic, present in nuclear RNA foci and correlated with a plethora of RNA splicing defects. Cardinal features of DM1 are myotonia and cardiac conduction abnormalities. Using transgenic mice, we have demonstrated that expression of the mutant DMPK 3'UTR is sufficient to elicit these features of DM1. Here, using these mice, we present a study of systemic treatment with an antisense oligonucleotide (ASO) (ISIS 486178) targeted to a non-CUG sequence within the 3'UTR of DMPK. RNA foci and DMPK 3'UTR mRNA levels were reduced in both the heart and skeletal muscles. This correlated with improvements in several splicing defects in skeletal and cardiac muscles. The treatment reduced myotonia and this correlated with increased Clcn1 expression. Furthermore, functional testing showed improvements in treadmill running. Of note, we demonstrate that the ASO treatment reversed the cardiac conduction abnormalities, and this correlated with restoration of Gja5 (connexin 40) expression in the heart. This is the first time that an ASO targeting a non-CUG sequence within the DMPK 3'UTR has demonstrated benefit on the key DM1 phenotypes of myotonia and cardiac conduction defects. Our data also shows for the first time that ASOs may be a viable option for treating cardiac pathology in DM1.
Collapse
Affiliation(s)
- Ramesh S Yadava
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Qing Yu
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Mahua Mandal
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Frank Rigo
- Ionis Pharmaceuticals Inc., Carlsbad, CA 90210, USA
| | | | - Mani S Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
16
|
Misra C, Bangru S, Lin F, Lam K, Koenig SN, Lubbers ER, Hedhli J, Murphy NP, Parker DJ, Dobrucki LW, Cooper TA, Tajkhorshid E, Mohler PJ, Kalsotra A. Aberrant Expression of a Non-muscle RBFOX2 Isoform Triggers Cardiac Conduction Defects in Myotonic Dystrophy. Dev Cell 2020; 52:748-763.e6. [PMID: 32109384 PMCID: PMC7098852 DOI: 10.1016/j.devcel.2020.01.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic genetic disorder caused by the CTG repeat expansion in the 3'-untranslated region of DMPK gene. Heart dysfunctions occur in ∼80% of DM1 patients and are the second leading cause of DM1-related deaths. Herein, we report that upregulation of a non-muscle splice isoform of RNA-binding protein RBFOX2 in DM1 heart tissue-due to altered splicing factor and microRNA activities-induces cardiac conduction defects in DM1 individuals. Mice engineered to express the non-muscle RBFOX240 isoform in heart via tetracycline-inducible transgenesis, or CRISPR/Cas9-mediated genome editing, reproduced DM1-related cardiac conduction delay and spontaneous episodes of arrhythmia. Further, by integrating RNA binding with cardiac transcriptome datasets from DM1 patients and mice expressing the non-muscle RBFOX2 isoform, we identified RBFOX240-driven splicing defects in voltage-gated sodium and potassium channels, which alter their electrophysiological properties. Thus, our results uncover a trans-dominant role for an aberrantly expressed RBFOX240 isoform in DM1 cardiac pathogenesis.
Collapse
Affiliation(s)
- Chaitali Misra
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Sushant Bangru
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Feikai Lin
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Kin Lam
- Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Sara N Koenig
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Ellen R Lubbers
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jamila Hedhli
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Nathaniel P Murphy
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Darren J Parker
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Lawrence W Dobrucki
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Thomas A Cooper
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Emad Tajkhorshid
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Peter J Mohler
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
17
|
McNally EM, Mann DL, Pinto Y, Bhakta D, Tomaselli G, Nazarian S, Groh WJ, Tamura T, Duboc D, Itoh H, Hellerstein L, Mammen PPA. Clinical Care Recommendations for Cardiologists Treating Adults With Myotonic Dystrophy. J Am Heart Assoc 2020; 9:e014006. [PMID: 32067592 PMCID: PMC7070199 DOI: 10.1161/jaha.119.014006] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myotonic dystrophy is an inherited systemic disorder affecting skeletal muscle and the heart. Genetic testing for myotonic dystrophy is diagnostic and identifies those at risk for cardiac complications. The 2 major genetic forms of myotonic dystrophy, type 1 and type 2, differ in genetic etiology yet share clinical features. The cardiac management of myotonic dystrophy should include surveillance for arrhythmias and left ventricular dysfunction, both of which occur in progressive manner and contribute to morbidity and mortality. To promote the development of care guidelines for myotonic dystrophy, the Myotonic Foundation solicited the input of care experts and organized the drafting of these recommendations. As a rare disorder, large scale clinical trial data to guide the management of myotonic dystrophy are largely lacking. The following recommendations represent expert consensus opinion from those with experience in the management of myotonic dystrophy, in part supported by literature-based evidence where available.
Collapse
Affiliation(s)
| | | | - Yigal Pinto
- University of AmsterdamAmsterdamThe Netherlands
| | | | | | | | | | - Takuhisa Tamura
- National Hospital Organization Higashisaitama National HospitalSaitamaJapan
| | - Denis Duboc
- Hopital CochinUniversite Paris DescartesParisFrance
| | - Hideki Itoh
- Shiga University of Medical ScienceShigaJapan
| | | | | |
Collapse
|
18
|
Asatryan B, Medeiros-Domingo A. Molecular and genetic insights into progressive cardiac conduction disease. Europace 2019; 21:1145-1158. [DOI: 10.1093/europace/euz109] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
Abstract
Progressive cardiac conduction disease (PCCD) is often a primarily genetic disorder, with clinical and genetic overlaps with other inherited cardiac and metabolic diseases. A number of genes have been implicated in PCCD pathogenesis with or without structural heart disease or systemic manifestations. Precise genetic diagnosis contributes to risk stratification, better selection of specific therapy and allows familiar cascade screening. Cardiologists should be aware of the different phenotypes emerging from different gene-mutations and the potential risk of sudden cardiac death. Genetic forms of PCCD often overlap or coexist with other inherited heart diseases or manifest in the context of multisystem syndromes. Despite the significant advances in the knowledge of the genetic architecture of PCCD and overlapping diseases, in a measurable fraction of PCCD cases, including in familial clustering of disease, investigations of known cardiac disease-associated genes fail to reveal the underlying substrate, suggesting that new causal genes are yet to be discovered. Here, we provide insight into genetics and molecular mechanisms of PCCD and related diseases. We also highlight the phenotypic overlaps of PCCD with other inherited cardiac and metabolic diseases, present unmet challenges in clinical practice, and summarize the available therapeutic options for affected patients.
Collapse
Affiliation(s)
- Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, Freiburgstrasse 8, Bern, Switzerland
| | | |
Collapse
|
19
|
Non-invasive monitoring of alternative splicing outcomes to identify candidate therapies for myotonic dystrophy type 1. Nat Commun 2018; 9:5227. [PMID: 30531949 PMCID: PMC6286378 DOI: 10.1038/s41467-018-07517-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 11/07/2018] [Indexed: 01/16/2023] Open
Abstract
During drug development, tissue samples serve as indicators of disease activity and pharmacodynamic responses. Reliable non-invasive measures of drug target engagement will facilitate identification of promising new treatments. Here we develop and validate a novel bi-transgenic mouse model of myotonic dystrophy type 1 (DM1) in which expression of either DsRed or GFP is determined by alternative splicing of an upstream minigene that is mis-regulated in DM1. Using a novel in vivo fluorescence spectroscopy system, we show that quantitation of the DsRed/GFP ratio provides an accurate estimation of splicing outcomes in muscle tissue of live mice that nearly doubles throughput over conventional fluorescence imaging techniques. Serial in vivo spectroscopy measurements in mice treated with a C16 fatty acid ligand conjugated antisense (LICA) oligonucleotide reveal a dose-dependent therapeutic response within seven days, confirm a several-week duration of action, and demonstrate a two-fold greater target engagement as compared to the unconjugated parent oligonucleotide.
Collapse
|
20
|
Picchio L, Legagneux V, Deschamps S, Renaud Y, Chauveau S, Paillard L, Jagla K. Bruno-3 regulates sarcomere component expression and contributes to muscle phenotypes of myotonic dystrophy type 1. Dis Model Mech 2018; 11:dmm.031849. [PMID: 29716962 PMCID: PMC5992612 DOI: 10.1242/dmm.031849] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 04/18/2018] [Indexed: 01/22/2023] Open
Abstract
Steinert disease, or myotonic dystrophy type 1 (DM1), is a multisystemic disorder caused by toxic noncoding CUG repeat transcripts, leading to altered levels of two RNA binding factors, MBNL1 and CELF1. The contribution of CELF1 to DM1 phenotypes is controversial. Here, we show that the Drosophila CELF1 family member, Bru-3, contributes to pathogenic muscle defects observed in a Drosophila model of DM1. Bru-3 displays predominantly cytoplasmic expression in muscles and its muscle-specific overexpression causes a range of phenotypes also observed in the fly DM1 model, including affected motility, fiber splitting, reduced myofiber length and altered myoblast fusion. Interestingly, comparative genome-wide transcriptomic analyses revealed that Bru-3 negatively regulates levels of mRNAs encoding a set of sarcomere components, including Actn transcripts. Conversely, it acts as a positive regulator of Actn translation. As CELF1 displays predominantly cytoplasmic expression in differentiating C2C12 myotubes and binds to Actn mRNA, we hypothesize that it might exert analogous functions in vertebrate muscles. Altogether, we propose that cytoplasmic Bru-3 contributes to DM1 pathogenesis in a Drosophila model by regulating sarcomeric transcripts and protein levels.
Collapse
Affiliation(s)
- Lucie Picchio
- GReD (Genetics, Reproduction and Development Laboratory), INSERM 1103, CNRS 6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Vincent Legagneux
- IGDR (Institut de Génétique et Développement de Rennes), UMR 6290 CNRS, Université de Rennes, 2 Avenue Léon Bernard, 35000 Rennes, France.,Inserm UMR1085 IRSET, Université de Rennes 1, 35000 Rennes, France.,CNRS-Université de Rennes1-INRIA, UMR6074 IRISA, 35000 Rennes, France
| | - Stephane Deschamps
- IGDR (Institut de Génétique et Développement de Rennes), UMR 6290 CNRS, Université de Rennes, 2 Avenue Léon Bernard, 35000 Rennes, France
| | - Yoan Renaud
- GReD (Genetics, Reproduction and Development Laboratory), INSERM 1103, CNRS 6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Sabine Chauveau
- GReD (Genetics, Reproduction and Development Laboratory), INSERM 1103, CNRS 6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Luc Paillard
- IGDR (Institut de Génétique et Développement de Rennes), UMR 6290 CNRS, Université de Rennes, 2 Avenue Léon Bernard, 35000 Rennes, France
| | - Krzysztof Jagla
- GReD (Genetics, Reproduction and Development Laboratory), INSERM 1103, CNRS 6293, University of Clermont Auvergne, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
21
|
Rinaldi C, Wood MJA. Antisense oligonucleotides: the next frontier for treatment of neurological disorders. Nat Rev Neurol 2017; 14:9-21. [PMID: 29192260 DOI: 10.1038/nrneurol.2017.148] [Citation(s) in RCA: 555] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antisense oligonucleotides (ASOs) were first discovered to influence RNA processing and modulate protein expression over two decades ago; however, progress translating these agents into the clinic has been hampered by inadequate target engagement, insufficient biological activity, and off-target toxic effects. Over the years, novel chemical modifications of ASOs have been employed to address these issues. These modifications, in combination with elucidation of the mechanism of action of ASOs and improved clinical trial design, have provided momentum for the translation of ASO-based strategies into therapies. Many neurological conditions lack an effective treatment; however, as research progressively disentangles the pathogenic mechanisms of these diseases, they provide an ideal platform to test ASO-based strategies. This steady progress reached a pinnacle in the past few years with approvals of ASOs for the treatment of spinal muscular atrophy and Duchenne muscular dystrophy, which represent landmarks in a field in which disease-modifying therapies were virtually non-existent. With the rapid development of improved next-generation ASOs toward clinical application, this technology now holds the potential to have a dramatic effect on the treatment of many neurological conditions in the near future.
Collapse
Affiliation(s)
- Carlo Rinaldi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford OX1 3QX, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
22
|
Cardiac involvement in myotonic dystrophy: The role of troponins and N-terminal pro B-type natriuretic peptide. Atherosclerosis 2017; 267:110-115. [PMID: 29121498 DOI: 10.1016/j.atherosclerosis.2017.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 09/19/2017] [Accepted: 10/18/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are dominant inherited muscular dystrophies with multiple systemic involvement, often producing cardiac injury. This study sought to determine the clinical significance of elevated high sensitivity cardiac troponin T and I (hs-cTnT and hs-cTnI), and N-terminal pro B-type natriuretic peptide (NT-pro-BNP) in this population. METHODS Sixty DM patients (35 men and 25 women; mean age: 45.1 years, range: 12-73 years) underwent clinical cardiac investigations and measurements of serum hs-cTnT, hs-cTnI, creatine kinase (CK), and NT-proBNP. Left ventricular (LV) ejection fraction (EF) was assessed by echocardiography. RESULTS Genetic analysis revealed that 46 of the 60 patients were DM1, and 14 DM2. Blood measurements showed persistent elevation of hs-cTnT and CK in 55/60 DM patients (91.73%). In contrast, hs-cTnI values were persistently normal throughout the study. Only 2 patients showed an EF <50%, being the overall range of this population between 40% and 79%. We found ECG abnormalities in 19 patients. Of these patients, 13 showed first or second-degree atrio ventricular (AV) blocks (PR interval ≥ 200 ms), 4 showed a left bundle branch block (LBBB) prolonged (QRS duration ≥120 ms), and 2 had an incomplete bundle branch block (QRS duration between 110 and 119 ms). After excluding patients with EF <50%, NT-pro-BNP measurement > 125 pg/mL was an independent predictor of ECG abnormalities. CONCLUSIONS NT-pro-BNP levels may be considered to be used clinically to identify DM patients at increased risk of developing myocardial conduction abnormalities.
Collapse
|
23
|
Co-option of the cardiac transcription factor Nkx2.5 during development of the emu wing. Nat Commun 2017; 8:132. [PMID: 28743862 PMCID: PMC5526984 DOI: 10.1038/s41467-017-00112-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/02/2017] [Indexed: 01/12/2023] Open
Abstract
The ratites are a distinctive clade of flightless birds, typified by the emu and ostrich that have acquired a range of unique anatomical characteristics since diverging from basal Aves at least 100 million years ago. The emu possesses a vestigial wing with a single digit and greatly reduced forelimb musculature. However, the embryological basis of wing reduction and other anatomical changes associated with loss of flight are unclear. Here we report a previously unknown co-option of the cardiac transcription factor Nkx2.5 to the forelimb in the emu embryo, but not in ostrich, or chicken and zebra finch, which have fully developed wings. Nkx2.5 is expressed in emu limb bud mesenchyme and maturing wing muscle, and mis-expression of Nkx2.5 throughout the limb bud in chick results in wing reductions. We propose that Nkx2.5 functions to inhibit early limb bud expansion and later muscle growth during development of the vestigial emu wing. The transcription factor Nkx2.5 is essential for heart development. Here, the authors identify a previously unknown expression domain for Nkx2.5 in the emu wing and explore its role in diminished wing bud development in the flightless emu, compared with three other birds that have functional wings.
Collapse
|
24
|
Valaperta R, Gaeta M, Cardani R, Lombardi F, Rampoldi B, De Siena C, Mori F, Fossati B, Gaia P, Ferraro OE, Villani S, Iachettini S, Piccoli M, Cirillo F, Pusineri E, Meola G, Costa E. High-sensitive cardiac troponin T (hs-cTnT) assay as serum biomarker to predict cardiac risk in myotonic dystrophy: A case-control study. Clin Chim Acta 2016; 463:122-128. [DOI: 10.1016/j.cca.2016.10.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/21/2016] [Indexed: 10/20/2022]
|
25
|
Kim YK, Yadava RS, Mandal M, Mahadevan K, Yu Q, Leitges M, Mahadevan MS. Disease Phenotypes in a Mouse Model of RNA Toxicity Are Independent of Protein Kinase Cα and Protein Kinase Cβ. PLoS One 2016; 11:e0163325. [PMID: 27657532 PMCID: PMC5033491 DOI: 10.1371/journal.pone.0163325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023] Open
Abstract
Myotonic dystrophy type 1(DM1) is the prototype for diseases caused by RNA toxicity. RNAs from the mutant allele contain an expanded (CUG)n tract within the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. The toxic RNAs affect the function of RNA binding proteins leading to sequestration of muscleblind-like (MBNL) proteins and increased levels of CELF1 (CUGBP, Elav-like family member 1). The mechanism for increased CELF1 is not very clear. One favored proposition is hyper-phosphorylation of CELF1 by Protein Kinase C alpha (PKCα) leading to increased CELF1 stability. However, most of the evidence supporting a role for PKC-α relies on pharmacological inhibition of PKC. To further investigate the role of PKCs in the pathogenesis of RNA toxicity, we generated transgenic mice with RNA toxicity that lacked both the PKCα and PKCβ isoforms. We find that these mice show similar disease progression as mice wildtype for the PKC isoforms. Additionally, the expression of CELF1 is also not affected by deficiency of PKCα and PKCβ in these RNA toxicity mice. These data suggest that disease phenotypes of these RNA toxicity mice are independent of PKCα and PKCβ.
Collapse
Affiliation(s)
- Yun K. Kim
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ramesh S. Yadava
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Mahua Mandal
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Karunasai Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Qing Yu
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael Leitges
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| | - Mani S. Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
26
|
Splicing misregulation of SCN5A contributes to cardiac-conduction delay and heart arrhythmia in myotonic dystrophy. Nat Commun 2016; 7:11067. [PMID: 27063795 PMCID: PMC4831019 DOI: 10.1038/ncomms11067] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 02/16/2016] [Indexed: 12/11/2022] Open
Abstract
Myotonic dystrophy (DM) is caused by the expression of mutant RNAs containing expanded CUG repeats that sequester muscleblind-like (MBNL) proteins, leading to alternative splicing changes. Cardiac alterations, characterized by conduction delays and arrhythmia, are the second most common cause of death in DM. Using RNA sequencing, here we identify novel splicing alterations in DM heart samples, including a switch from adult exon 6B towards fetal exon 6A in the cardiac sodium channel, SCN5A. We find that MBNL1 regulates alternative splicing of SCN5A mRNA and that the splicing variant of SCN5A produced in DM presents a reduced excitability compared with the control adult isoform. Importantly, reproducing splicing alteration of Scn5a in mice is sufficient to promote heart arrhythmia and cardiac-conduction delay, two predominant features of myotonic dystrophy. In conclusion, misregulation of the alternative splicing of SCN5A may contribute to a subset of the cardiac dysfunctions observed in myotonic dystrophy. Patients with myotonic dystrophy (MD) suffer from severe cardiac issues of unknown aetiology. Freyermuth et al. show that fatal changes in cardiac electrophysiological properties in humans and mice with MD may arise from misregulation of the alternative splicing of the cardiac Na+ channel SCN5A transcript, resulting in expression of its fetal form.
Collapse
|
27
|
Yadava RS, Foff EP, Yu Q, Gladman JT, Zheng TS, Mahadevan MS. TWEAK Regulates Muscle Functions in a Mouse Model of RNA Toxicity. PLoS One 2016; 11:e0150192. [PMID: 26901467 PMCID: PMC4762946 DOI: 10.1371/journal.pone.0150192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 02/09/2016] [Indexed: 12/31/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most common form of muscular dystrophy in adults, is caused by toxic RNAs produced from the mutant DM protein kinase (DMPK) gene. DM1 is characterized by progressive muscle wasting and weakness. Therapeutic strategies have mainly focused on targeting the toxic RNA. Previously, we found that fibroblast growth factor-inducible 14 (Fn14), the receptor for TWEAK, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and that blocking TWEAK/Fn14 signaling improves muscle function and histology. Here, we studied the effect of Tweak deficiency in a RNA toxicity mouse model. The genetic deletion of Tweak in these mice significantly reduced muscle damage and improved muscle function. In contrast, administration of TWEAK in the RNA toxicity mice impaired functional outcomes and worsened muscle histopathology. These studies show that signaling via TWEAK is deleterious to muscle in RNA toxicity and support the demonstrated utility of anti-TWEAK therapeutics.
Collapse
Affiliation(s)
- Ramesh S. Yadava
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
| | - Erin P. Foff
- Department of Neurology, University of Virginia, Charlottesville, VA, United States of America
| | - Qing Yu
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
| | - Jordan T. Gladman
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
| | - Timothy S. Zheng
- Department of Immunology, Biogen Idec, Cambridge, MA, United States of America
| | - Mani S. Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
- * E-mail:
| |
Collapse
|
28
|
Di Salvo TG, Guo Y, Su YR, Clark T, Brittain E, Absi T, Maltais S, Hemnes A. Right ventricular long noncoding RNA expression in human heart failure. Pulm Circ 2015; 5:135-61. [PMID: 25992278 PMCID: PMC4405709 DOI: 10.1086/679721] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 08/13/2014] [Indexed: 12/11/2022] Open
Abstract
The expression of long noncoding RNAs (lncRNAs) in human heart failure (HF) has not been widely studied. Using RNA sequencing (RNA-Seq), we compared lncRNA expression in 22 explanted human HF hearts with lncRNA expression in 5 unused donor human hearts. We used Cufflinks to identify isoforms and DESeq to identify differentially expressed genes. We identified the noncoding RNAs by cross-reference to Ensembl release 73 (Genome Reference Consortium human genome build 37) and explored possible functional roles using a variety of online tools. In HF hearts, RNA-Seq identified 84,793 total messenger RNA coding and noncoding different transcripts, including 13,019 protein-coding genes, 2,085 total lncRNA genes, and 1,064 pseudogenes. By Ensembl noncoding RNA categories, there were 48 lncRNAs, 27 pseudogenes, and 30 antisense RNAs for a total of 105 differentially expressed lncRNAs in HF hearts. Compared with donor hearts, HF hearts exhibited differential expression of 7.7% of protein-coding genes, 3.7% of lncRNAs (including pseudogenes), and 2.5% of pseudogenes. There were not consistent correlations between antisense lncRNAs and parent genes and between pseudogenes and parent genes, implying differential regulation of expression. Exploratory in silico functional analyses using online tools suggested a variety of possible lncRNA regulatory roles. By providing a comprehensive profile of right ventricular polyadenylated messenger RNA transcriptome in HF, RNA-Seq provides an inventory of differentially expressed lncRNAs, including antisense transcripts and pseudogenes, for future mechanistic study.
Collapse
Affiliation(s)
- Thomas G. Di Salvo
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Nashville, Tennessee, USA
| | - Yan Guo
- Vanderbilt Medical Center VANTAGE Core Laboratory, Nashville, Tennessee, USA
| | - Yan Ru Su
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Nashville, Tennessee, USA
| | - Travis Clark
- Vanderbilt Medical Center VANTAGE Core Laboratory, Nashville, Tennessee, USA
| | - Evan Brittain
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Nashville, Tennessee, USA
| | - Tarek Absi
- Division of Cardiovascular Surgery, Vanderbilt Heart and Vascular Institute, Nashville, Tennessee, USA
| | - Simon Maltais
- Division of Cardiovascular Surgery, Vanderbilt Heart and Vascular Institute, Nashville, Tennessee, USA
| | - Anna Hemnes
- Pulmonary and Critical Care Medicine, Vanderbilt Department of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
29
|
Chau A, Kalsotra A. Developmental insights into the pathology of and therapeutic strategies for DM1: Back to the basics. Dev Dyn 2015; 244:377-90. [PMID: 25504326 DOI: 10.1002/dvdy.24240] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/25/2014] [Accepted: 11/27/2014] [Indexed: 12/25/2022] Open
Abstract
Myotonic Dystrophy type 1 (DM1), the most prevalent adult onset muscular dystrophy, is a trinucleotide repeat expansion disease caused by CTG expansion in the 3'-UTR of DMPK gene. This expansion results in the expression of toxic gain-of-function RNA that forms ribonuclear foci and disrupts normal activities of RNA-binding proteins belonging to the MBNL and CELF families. Changes in alternative splicing, translation, localization, and mRNA stability due to sequestration of MBNL proteins and up-regulation of CELF1 are key to DM1 pathology. However, recent discoveries indicate that pathogenic mechanisms of DM1 involves many other factors as well, including repeat associated translation, activation of PKC-dependent signaling pathway, aberrant polyadenylation, and microRNA deregulation. Expression of the toxic repeat RNA culminates in the developmental remodeling of the transcriptome, which produces fetal isoforms of proteins that are unable to fulfill the physiological requirements of adult tissues. This review will describe advances in the understanding of DM1 pathogenesis as well as current therapeutic developments for DM1.
Collapse
Affiliation(s)
- Anthony Chau
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Illinois; Department of Medical Biochemistry, University of Illinois, Urbana-Champaign, Illinois; Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Illinois
| | | |
Collapse
|
30
|
Yadava RS, Foff EP, Yu Q, Gladman JT, Kim YK, Bhatt KS, Thornton CA, Zheng TS, Mahadevan MS. TWEAK/Fn14, a pathway and novel therapeutic target in myotonic dystrophy. Hum Mol Genet 2014; 24:2035-48. [PMID: 25504044 DOI: 10.1093/hmg/ddu617] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most prevalent muscular dystrophy in adults, is characterized by progressive muscle wasting and multi-systemic complications. DM1 is the prototype for disorders caused by RNA toxicity. Currently, no therapies exist. Here, we identify that fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor receptor super-family, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and in tissues from DM1 patients, and that its expression correlates with severity of muscle pathology. This is associated with downstream signaling through the NF-κB pathways. In mice with RNA toxicity, genetic deletion of Fn14 results in reduced muscle pathology and better function. Importantly, blocking TWEAK/Fn14 signaling with an anti-TWEAK antibody likewise improves muscle histopathology and functional outcomes in affected mice. These results reveal new avenues for therapeutic development and provide proof of concept for a novel therapeutic target for which clinically available therapy exists to potentially treat muscular dystrophy in DM1.
Collapse
Affiliation(s)
| | - Erin P Foff
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | - Kirti S Bhatt
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA and
| | - Charles A Thornton
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA and
| | - Timothy S Zheng
- Department of Immunology, Biogen Idec, Cambridge, MA 02142, USA
| | | |
Collapse
|
31
|
Affiliation(s)
- Thomas G. Di Salvo
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Nashville TN
| | - Saptarsi M. Haldar
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH
- Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH
| |
Collapse
|
32
|
Gladman JT, Yadava RS, Mandal M, Yu Q, Kim YK, Mahadevan MS. NKX2-5, a modifier of skeletal muscle pathology due to RNA toxicity. Hum Mol Genet 2014; 24:251-64. [PMID: 25168381 DOI: 10.1093/hmg/ddu443] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
RNA toxicity is implicated in a number of disorders; especially those associated with expanded repeat sequences, such as myotonic dystrophy (DM1). Previously, we have shown increased NKX2-5 expression in RNA toxicity associated with DM1. Here, we investigate the relationship between NKX2-5 expression and muscle pathology due to RNA toxicity. In skeletal muscle from mice with RNA toxicity and individuals with DM1, expression of Nkx2-5 or NKX2-5 and its downstream targets are significantly correlated with severity of histopathology. Using C2C12 myoblasts, we show that over-expression of NKX2-5 or mutant DMPK 3'UTR results in myogenic differentiation defects, which can be rescued by knockdown of Nkx2-5, despite continued toxic RNA expression. Furthermore, in a mouse model of NKX2-5 over-expression, we find defects in muscle regeneration after induced damage, similar to those seen in mice with RNA toxicity. Using mouse models of Nkx2-5 over-expression and depletion, we find that NKX2-5 levels modify disease phenotypes in mice with RNA toxicity.
Collapse
Affiliation(s)
- Jordan T Gladman
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ramesh S Yadava
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Mahua Mandal
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Qing Yu
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yun K Kim
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Mani S Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
33
|
Garcia SMDA, Tabach Y, Lourenço GF, Armakola M, Ruvkun G. Identification of genes in toxicity pathways of trinucleotide-repeat RNA in C. elegans. Nat Struct Mol Biol 2014; 21:712-20. [PMID: 25038802 PMCID: PMC4125460 DOI: 10.1038/nsmb.2858] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 06/18/2014] [Indexed: 01/15/2023]
Abstract
Myotonic dystrophy disorders are caused by expanded CUG repeats in non-coding regions. To reveal mechanisms of CUG repeat pathogenesis we used C. elegans expressing CUG repeats to identify gene inactivations that modulate CUG repeat toxicity. We identified 15 conserved genes that function as suppressors or enhancers of CUG repeat-induced toxicity and modulate formation of nuclear RNA foci by CUG repeats. These genes regulated CUG repeat-induced toxicity through distinct mechanisms including RNA export and RNA clearance, suggesting that CUG repeat toxicity is mediated by multiple pathways. A subset is shared with other degenerative disorders. The nonsense-mediated mRNA decay (NMD) pathway plays a conserved role regulating CUG repeat RNA transcript levels and toxicity, and NMD recognition of toxic RNAs depends on 3′UTR GC nucleotide content. Our studies suggest a broader surveillance role for NMD where variations in this pathway influence multiple degenerative diseases.
Collapse
Affiliation(s)
- Susana M D A Garcia
- 1] Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuval Tabach
- 1] Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Guinevere F Lourenço
- 1] Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA. [3]
| | - Maria Armakola
- 1] Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Gary Ruvkun
- 1] Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Li X, Wu Z, Fu X, Han W. lncRNAs: insights into their function and mechanics in underlying disorders. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 762:1-21. [PMID: 25485593 DOI: 10.1016/j.mrrev.2014.04.002] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 12/14/2022]
Abstract
Genomes of complex organisms are characterized by the pervasive expression of different types of noncoding RNAs (ncRNAs). lncRNAs constitute a large family of long—arbitrarily defined as being longer than 200 nucleotides—ncRNAs that are expressed throughout the cell and that include thousands of different species. While these new and enigmatic players in the complex transcriptional milieu are encoded by a significant proportion of the genome, their functions are mostly unknown at present. Existing examples suggest that lncRNAs have fulfilled a wide variety of regulatory roles at almost every stage of gene expression. These roles, which encompass signal, decoy, scaffold and guide capacities, derive from folded modular domains in lncRNAs. Early discoveries support a paradigm in which lncRNAs regulate transcription networks via chromatin modulation, but new functions are steadily emerging. Given the biochemical versatility of RNA, lncRNAs may be used for various tasks, including posttranscriptional processing. In addition, long intergenic ncRNAs (lincRNAs) are strongly enriched for trait-associated SNPs, which suggest a new mechanism by which intergenic trait-associated regions might function. Moreover, multiple lines of evidence increasingly link mutations and dysregulations of lncRNAs to diverse human diseases, especially disorders related to aging. In this article, we review the current state of the knowledge of the lncRNA field, discussing what is known about the genomic contexts, biological functions and mechanisms of action of these molecules. We highlight the growing evidence for the importance of lncRNAs in diverse human disorders and the indications that their dysregulations and mutations underlie some aging-related disorders. Finally, we consider the potential medical implications, and future potential in the application of lncRNAs as therapeutic targets and diagnostic markers.
Collapse
Affiliation(s)
- Xiaolei Li
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhiqiang Wu
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaobing Fu
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China; Key Laboratory of Wound Healing and Cell Biology, Institute of Burns, The First Affiliated Hospital to the Chinese PLA General Hospital, Trauma Center of Postgraduate Medical School, Beijing 100037, China.
| | - Weidong Han
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
35
|
Schluesener JK, Schluesener H. Plant polyphenols in the treatment of age-associated diseases: revealing the pleiotropic effects of icariin by network analysis. Mol Nutr Food Res 2013; 58:49-60. [PMID: 24311544 DOI: 10.1002/mnfr.201300409] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/18/2013] [Accepted: 10/30/2013] [Indexed: 12/15/2022]
Abstract
Polyphenols are a broad class of compounds. Some are ingested in substantial quantities from nutritional sources, more are produced by medicinal plants, and some of them are taken as drugs. It is becoming clear, that a single polyphenol is impacting several cellular pathways. Thus, a network approach is becoming feasible, describing the interaction of a single polyphenol with cellular networks. Here we have selected icariin to draw a prototypic network of icariin activities. Icariin appears to be a promising drug to treat major age-related diseases, like neurodegeneration, memory and depressive disorders, chronic inflammation, diabetes, and osteoporosis. It interacts with several relevant pathways, like PDE, TGF-ß, MAPK, PPAR, NOS, IGF, Sirtuin, and others. Such networks will be useful to future comparative studies of complex effects of polyphenols.
Collapse
Affiliation(s)
- Jan Kevin Schluesener
- Division of Immunopathology of the Nervous System, Department of Neuropathology, Institute of Pathology and Neuropathology, University of Tuebingen, Tuebingen, Germany
| | | |
Collapse
|
36
|
Brugada syndrome and abnormal splicing of SCN5A in myotonic dystrophy type 1. Arch Cardiovasc Dis 2013; 106:635-43. [DOI: 10.1016/j.acvd.2013.08.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/16/2013] [Accepted: 08/29/2013] [Indexed: 01/22/2023]
|
37
|
Age of onset of RNA toxicity influences phenotypic severity: evidence from an inducible mouse model of myotonic dystrophy (DM1). PLoS One 2013; 8:e72907. [PMID: 24039817 PMCID: PMC3764231 DOI: 10.1371/journal.pone.0072907] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/13/2013] [Indexed: 01/23/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy in adults. It is caused by an expanded (CTG)n tract in the 3′ UTR of the Dystrophia Myotonica Protein Kinase (DMPK) gene. This causes nuclear retention of the mutant mRNA into ribonuclear foci and sequestration of interacting RNA-binding proteins (such as muscleblind-like 1 (MBNL1)). More severe congenital and childhood-onset forms of the disease exist but are less understood than the adult disease, due in part to the lack of adequate animal models. To address this, we utilized transgenic mice over-expressing the DMPK 3′ UTR as part of an inducible RNA transcript to model early-onset myotonic dystrophy. In mice in which transgene expression was induced during embryogenesis, we found that by two weeks after birth, mice reproduced cardinal features of myotonic dystrophy, including myotonia, cardiac conduction abnormalities, muscle weakness, histopathology and mRNA splicing defects. Notably, these defects were more severe than in adult mice induced for an equivalent period of exposure to RNA toxicity. Additionally, the utility of the model was tested by over-expressing MBNL1, a key therapeutic strategy being actively pursued for treating the disease phenotypes associated with DM1. Significantly, increased MBNL1 in skeletal muscle partially corrected myotonia and splicing defects present in these mice, demonstrating the responsiveness of the model to relevant therapeutic interventions. Furthermore, these results also represent the first murine model for early-onset DM1 and provide a tool to investigate the effects of RNA toxicity at various stages of development.
Collapse
|
38
|
Kim YK, Mandal M, Yadava RS, Paillard L, Mahadevan MS. Evaluating the effects of CELF1 deficiency in a mouse model of RNA toxicity. Hum Mol Genet 2013; 23:293-302. [PMID: 24001600 DOI: 10.1093/hmg/ddt419] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most common form of adult-onset muscular dystrophy, is caused by an expanded (CTG)n repeat in the 3' untranslated region of the DM protein kinase (DMPK) gene. The toxic RNA transcripts produced from the mutant allele alter the function of RNA-binding proteins leading to the functional depletion of muscleblind-like (MBNL) proteins and an increase in steady state levels of CUG-BP1 (CUGBP-ETR-3 like factor 1, CELF1). The role of increased CELF1 in DM1 pathogenesis is well studied using genetically engineered mouse models. Also, as a potential therapeutic strategy, the benefits of increasing MBNL1 expression have recently been reported. However, the effect of reduction of CELF1 is not yet clear. In this study, we generated CELF1 knockout mice, which also carry an inducible toxic RNA transgene to test the effects of CELF1 reduction in RNA toxicity. We found that the absence of CELF1 did not correct splicing defects. It did however mitigate the increase in translational targets of CELF1 (MEF2A and C/EBPβ). Notably, we found that loss of CELF1 prevented deterioration of muscle function by the toxic RNA, and resulted in better muscle histopathology. These data suggest that while reduction of CELF1 may be of limited benefit with respect to DM1-associated spliceopathy, it may be beneficial to the muscular dystrophy associated with RNA toxicity.
Collapse
Affiliation(s)
- Yun Kyoung Kim
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
39
|
Gauthier M, Marteyn A, Denis JA, Cailleret M, Giraud-Triboult K, Aubert S, Lecuyer C, Marie J, Furling D, Vernet R, Yanguas C, Baldeschi C, Pietu G, Peschanski M, Martinat C. A defective Krab-domain zinc-finger transcription factor contributes to altered myogenesis in myotonic dystrophy type 1. Hum Mol Genet 2013; 22:5188-98. [PMID: 23922231 DOI: 10.1093/hmg/ddt373] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an RNA-mediated disorder caused by a non-coding CTG repeat expansion that, in particular, provokes functional alteration of CUG-binding proteins. As a consequence, several genes with misregulated alternative splicing have been linked to clinical symptoms. In our search for additional molecular mechanisms that would trigger functional defects in DM1, we took advantage of mutant gene-carrying human embryonic stem cell lines to identify differentially expressed genes. Among the different genes found to be misregulated by DM1 mutation, one strongly downregulated gene encodes a transcription factor, ZNF37A. In this paper, we show that this defect in expression, which derives from a loss of RNA stability, is controlled by the RNA-binding protein, CUGBP1, and is associated with impaired myogenesis-a functional defect reminiscent of that observed in DM1. Loss of the ZNF37A protein results in changes in the expression of the subunit α1 of the receptor for the interleukin 13. This suggests that the pathological molecular mechanisms linking ZNF37A and myogenesis may involve the signaling pathway that is known to promote myoblast recruitment during development and regeneration.
Collapse
|
40
|
The alternative heart: impact of alternative splicing in heart disease. J Cardiovasc Transl Res 2013; 6:945-55. [PMID: 23775418 DOI: 10.1007/s12265-013-9482-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/04/2013] [Indexed: 01/16/2023]
Abstract
Alternative splicing is the main driver of protein diversity and allows the production of different proteins from each gene in the genome. Changes in exon exclusion, intron retention or the use of alternative splice sites can alter protein structure, localisation, regulation and function. In the heart, alternative splicing of sarcomeric genes, ion channels and cell signalling proteins can lead to cardiomyopathies, arrhythmias and other pathologies. Also, a number of inherited conditions and heart-related diseases develop as a result of mutations affecting splicing. Here, we review the impact that changes in alternative splicing have on individual genes and on whole biological processes associated with heart disease. We also discuss promising therapeutic tools based on the manipulation of alternative splicing.
Collapse
|
41
|
Costa MW, Guo G, Wolstein O, Vale M, Castro ML, Wang L, Otway R, Riek P, Cochrane N, Furtado M, Semsarian C, Weintraub RG, Yeoh T, Hayward C, Keogh A, Macdonald P, Feneley M, Graham RM, Seidman JG, Seidman CE, Rosenthal N, Fatkin D, Harvey RP. Functional characterization of a novel mutation in NKX2-5 associated with congenital heart disease and adult-onset cardiomyopathy. ACTA ACUST UNITED AC 2013; 6:238-47. [PMID: 23661673 DOI: 10.1161/circgenetics.113.000057] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The transcription factor NKX2-5 is crucial for heart development, and mutations in this gene have been implicated in diverse congenital heart diseases and conduction defects in mouse models and humans. Whether NKX2-5 mutations have a role in adult-onset heart disease is unknown. METHODS AND RESULTS Mutation screening was performed in 220 probands with adult-onset dilated cardiomyopathy. Six NKX2-5 coding sequence variants were identified, including 3 nonsynonymous variants. A novel heterozygous mutation, I184M, located within the NKX2-5 homeodomain, was identified in 1 family. A subset of family members had congenital heart disease, but there was an unexpectedly high prevalence of dilated cardiomyopathy. Functional analysis of I184M in vitro demonstrated a striking increase in protein expression when transfected into COS-7 cells or HL-1 cardiomyocytes because of reduced degradation by the Ubiquitin-proteasome system. In functional assays, DNA-binding activity of I184M was reduced, resulting in impaired activation of target genes despite increased expression levels of mutant protein. CONCLUSIONS Certain NKX2-5 homeodomain mutations show abnormal protein degradation via the Ubiquitin-proteasome system and partially impaired transcriptional activity. We propose that this class of mutation can impair heart development and mature heart function and contribute to NKX2-5-related cardiomyopathies with graded severity.
Collapse
Affiliation(s)
- Mauro W Costa
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sicot G, Gomes-Pereira M. RNA toxicity in human disease and animal models: from the uncovering of a new mechanism to the development of promising therapies. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1390-409. [PMID: 23500957 DOI: 10.1016/j.bbadis.2013.03.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/01/2013] [Accepted: 03/04/2013] [Indexed: 01/06/2023]
Abstract
Mutant ribonucleic acid (RNA) molecules can be toxic to the cell, causing human disease through trans-acting dominant mechanisms. RNA toxicity was first described in myotonic dystrophy type 1, a multisystemic disorder caused by the abnormal expansion of a non-coding trinucleotide repeat sequence. The development of multiple and complementary animal models of disease has greatly contributed to clarifying the complex disease pathways mediated by toxic RNA molecules. RNA toxicity is not limited to myotonic dystrophy and spreads to an increasing number of human conditions, which share some unifying pathogenic events mediated by toxic RNA accumulation and disruption of RNA-binding proteins. The remarkable progress in the dissection of disease pathobiology resulted in the rational design of molecular therapies, which have been successfully tested in animal models. Toxic RNA diseases, and in particular myotonic dystrophy, clearly illustrate the critical contribution of animal models of disease in translational research: from gene mutation to disease mechanisms, and ultimately to therapy development. This article is part of a Special Issue entitled: Animal Models of Disease.
Collapse
|
43
|
Denis JA, Gauthier M, Rachdi L, Aubert S, Giraud-Triboult K, Poydenot P, Benchoua A, Champon B, Maury Y, Baldeschi C, Scharfmann R, Piétu G, Peschanski M, Martinat C. mTOR-dependent proliferation defect in human ES-derived neural stem cells affected by myotonic dystrophy type 1. J Cell Sci 2013; 126:1763-72. [PMID: 23444380 DOI: 10.1242/jcs.116285] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Patients with myotonic dystrophy type 1 exhibit a diversity of symptoms that affect many different organs. Among these are cognitive dysfunctions, the origin of which has remained elusive, partly because of the difficulty in accessing neural cells. Here, we have taken advantage of pluripotent stem cell lines derived from embryos identified during a pre-implantation genetic diagnosis for mutant-gene carriers, to produce early neuronal cells. Functional characterization of these cells revealed reduced proliferative capacity and increased autophagy linked to mTOR signaling pathway alterations. Interestingly, loss of function of MBNL1, an RNA-binding protein whose function is defective in DM1 patients, resulted in alteration of mTOR signaling, whereas gain-of-function experiments rescued the phenotype. Collectively, these results provide a mechanism by which DM1 mutation might affect a major signaling pathway and highlight the pertinence of using pluripotent stem cells to study neuronal defects.
Collapse
Affiliation(s)
- Jérôme Alexandre Denis
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, 91030 Evry cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The myotonic dystrophies (DM1 and DM2) are the paradigm for RNA toxicity in disease pathogenesis. The emphasis of this review will be on recent developments and issues in understanding the pathogenesis of DM1 and how this is driving the accelerated pace of translational and therapeutic developments. RECENT FINDINGS RNA toxicity in myotonic dystrophy is now associated with bi-directional antisense transcription, dysregulation of microRNAs and potentially non-ATG-mediated translation of homopolymeric toxic proteins. The role of other RNA-binding proteins beyond MBNL1 and CUGBP1, such as Staufen 1 and DDX5, are being identified and studied with respect to their role in myotonic dystrophy. New functions for MBNL1 in miR-1 biogenesis might have a clinically relevant role in myotonic dystrophy cardiac conduction defects and pathology. Advances are being made in identifying and characterizing small molecules with the potential to disrupt CUG-MBNL1 interactions. SUMMARY Mechanisms of RNA toxicity are moving beyond a simplistic 'foci-centric' view of DM1 pathogenesis as a spliceopathy due to MBNL1 sequestration. Therapeutic development for myotonic dystrophy is moving rapidly with the development of antisense and small molecule therapies. Clinically, significant emphasis is being placed on biomarker discovery and outcome measures as an essential prelude to clinical trials.
Collapse
|
45
|
Huin V, Vasseur F, Schraen-Maschke S, Dhaenens CM, Devos P, Dupont K, Sergeant N, Buée L, Lacour A, Hofmann-Radvanyi H, Sablonnière B. MBNL1 gene variants as modifiers of disease severity in myotonic dystrophy type 1. J Neurol 2012; 260:998-1003. [DOI: 10.1007/s00415-012-6740-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 10/16/2012] [Accepted: 10/29/2012] [Indexed: 01/14/2023]
|
46
|
Abstract
Heart function requires sophisticated regulatory networks to orchestrate organ development, physiological responses, and environmental adaptation. Until recently, it was thought that these regulatory networks are composed solely of protein-mediated transcriptional control and signaling systems; consequently, it was thought that cardiac disease involves perturbation of these systems. However, it is becoming evident that RNA, long considered to function primarily as the platform for protein production, may in fact play a major role in most, if not all, aspects of gene regulation, especially the epigenetic processes that underpin organogenesis. These include not only well-validated classes of regulatory RNAs, such as microRNAs, but also tens of thousands of long noncoding RNAs that are differentially expressed across the entire genome of humans and other animals. Here, we review this emerging landscape, summarizing what is known about their functions and their role in cardiac biology, and provide a toolkit to assist in exploring this previously hidden layer of gene regulation that may underpin heart adaptation and complex heart diseases.
Collapse
Affiliation(s)
- Nicole Schonrock
- From the Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia (N.S., R.R.H.); St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia (N.S., R.P.H., J.S.M.); and Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia (J.S.M.)
| | - Richard P. Harvey
- From the Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia (N.S., R.R.H.); St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia (N.S., R.P.H., J.S.M.); and Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia (J.S.M.)
| | - John S. Mattick
- From the Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia (N.S., R.R.H.); St. Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia (N.S., R.P.H., J.S.M.); and Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia (J.S.M.)
| |
Collapse
|
47
|
Renoux AJ, Todd PK. Neurodegeneration the RNA way. Prog Neurobiol 2011; 97:173-89. [PMID: 22079416 DOI: 10.1016/j.pneurobio.2011.10.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 10/06/2011] [Accepted: 10/27/2011] [Indexed: 02/09/2023]
Abstract
The expression, processing, transport and activities of both coding and non-coding RNAs play critical roles in normal neuronal function and differentiation. Over the past decade, these same pathways have come under scrutiny as potential contributors to neurodegenerative disease. Here we focus broadly on the roles of RNA and RNA processing in neurodegeneration. We first discuss a set of "RNAopathies", where non-coding repeat expansions drive pathogenesis through a surprisingly diverse set of mechanisms. We next explore an emerging class of "RNA binding proteinopathies" where redistribution and aggregation of the RNA binding proteins TDP-43 or FUS contribute to a potentially broad range of neurodegenerative disorders. Lastly, we delve into the potential contributions of alterations in both short and long non-coding RNAs to neurodegenerative illness.
Collapse
Affiliation(s)
- Abigail J Renoux
- Department of Molecular and Integrative Physiology, University of Michigan, 4005 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
48
|
Harmon EB, Harmon ML, Larsen TD, Yang J, Glasford JW, Perryman MB. Myotonic dystrophy protein kinase is critical for nuclear envelope integrity. J Biol Chem 2011; 286:40296-306. [PMID: 21949239 DOI: 10.1074/jbc.m111.241455] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myotonic dystrophy 1 (DM1) is a multisystemic disease caused by a triplet nucleotide repeat expansion in the 3' untranslated region of the gene coding for myotonic dystrophy protein kinase (DMPK). DMPK is a nuclear envelope (NE) protein that promotes myogenic gene expression in skeletal myoblasts. Muscular dystrophy research has revealed the NE to be a key determinant of nuclear structure, gene regulation, and muscle function. To investigate the role of DMPK in NE stability, we analyzed DMPK expression in epithelial and myoblast cells. We found that DMPK localizes to the NE and coimmunoprecipitates with Lamin-A/C. Overexpression of DMPK in HeLa cells or C2C12 myoblasts disrupts Lamin-A/C and Lamin-B1 localization and causes nuclear fragmentation. Depletion of DMPK also disrupts NE lamina, showing that DMPK is required for NE stability. Our data demonstrate for the first time that DMPK is a critical component of the NE. These novel findings suggest that reduced DMPK may contribute to NE instability, a common mechanism of skeletal muscle wasting in muscular dystrophies.
Collapse
Affiliation(s)
- Erin B Harmon
- Cardiovascular Health Research Center, Sanford Research/USD, Sioux Falls, South Dakota 57104, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Sicot G, Gourdon G, Gomes-Pereira M. Myotonic dystrophy, when simple repeats reveal complex pathogenic entities: new findings and future challenges. Hum Mol Genet 2011; 20:R116-23. [PMID: 21821673 DOI: 10.1093/hmg/ddr343] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Expanded, non-coding RNAs can exhibit a deleterious gain-of-function causing human disease through abnormal interactions with RNA-binding proteins. Myotonic dystrophy (DM), the prototypical example of an RNA-dominant disorder, is mediated by trinucleotide repeat-containing transcripts that deregulate alternative splicing. Spliceopathy has therefore been a major focus of DM research. However, changes in gene expression, protein translation and micro-RNA metabolism may also contribute to disease pathology. The exciting finding of bidirectional transcription and non-conventional RNA translation of trinucleotide repeat sequences points to a new scenario, in which DM is not mediated by one single expanded RNA transcript, but involves multiple pathogenic elements and pathways. The study of the growing number of human diseases associated with toxic repeat-containing transcripts provides important insight into the understanding of the complex pathways of RNA toxicity. This review describes some of the recent advances in the understanding of the molecular mechanisms behind DM and other RNA-dominant disorders.
Collapse
Affiliation(s)
- Géraldine Sicot
- INSERM U781, Université Paris Descartes, Hôpital Necker Enfants Malades, 156 rue de Vaugirard, Paris Cedex 15, France
| | | | | |
Collapse
|
50
|
Abstract
The development of effective therapies for neuromuscular disorders such as Duchenne muscular dystrophy (DMD) is hampered by considerable challenges: skeletal muscle is the most abundant tissue in the body, and many neuromuscular disorders are multisystemic conditions. However, despite these barriers there has recently been substantial progress in the search for novel treatments. In particular, the use of antisense oligonucleotides, which are designed to target RNA and modulate pre-mRNA splicing to restore functional protein isoforms or directly inhibit the toxic effects of pathogenic RNAs, offers great promise and these approaches are now being tested in the clinic. Here, we review recent advances in the development of such antisense oligonucleotides and other promising novel approaches, including the induction of readthrough nonsense mutations.
Collapse
Affiliation(s)
- Francesco Muntoni
- UCL Institute of Child Health and Great Ormond Street Hospital, 30 Guildford Street, London WC1N 1EH, UK.
| | | |
Collapse
|