1
|
Pascual-Morena C, Cavero-Redondo I, Saz-Lara A, Martínez-García I, Visier-Alfonso ME, Martínez-Vizcaíno V. Intellectual Profile in Myotonic Dystrophy Type 1 and Its Association With Its Onset: A Systematic Review and Meta-Analysis. Pediatr Neurol 2024; 161:9-17. [PMID: 39232462 DOI: 10.1016/j.pediatrneurol.2024.08.002] [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] [Received: 02/21/2023] [Revised: 07/14/2024] [Accepted: 08/03/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is caused by mutations in the DMPK gene, and it is associated with cognitive deficits and intelligence below normative values. The objective of this systematic review and meta-analysis was to estimate the overall intelligence and proportion of intellectual development disorder (IDD) in the population with DM1 and its association with its onset. METHODS Systematic searches of Medline, Scopus, Web of Science, and Cochrane Library were performed from inception to January 2023. Studies that determined the full intelligence quotient (FIQ) or the IDD proportion in populations with DM1 were included. Meta-analyses of the FIQ and IDD and the FIQ mean difference and IDD prevalence ratios (PRs) by disease onset, inheritance, and genotype were conducted. RESULTS Forty-five studies were included in the meta-analyses, and all were performed in the DM1 population. The FIQ and IDD in DM1 were 77.90 (71.98, 83.81) and 0.44 (0.27, 0.60), respectively. Furthermore, DM1 onset was negatively associated with intelligence. Thus, the comparison "Congenital versus Adult" onsets resulted in an intelligence quotient of -41.61 (-47.81, -35.40) points and a PR of IDD of 9.49 (3.23, 27.89). Finally, maternal inheritance was also negatively associated, but the genotype did not have a statistically significant association. CONCLUSIONS The alterations in intelligence in DM1 are highly associated with the onset of the disease. However, the genotype did not explain these alterations well and there may be other genetic or epigenetic factors that should be considered.
Collapse
Affiliation(s)
- Carlos Pascual-Morena
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain; Faculty of Nursing, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Iván Cavero-Redondo
- CarVasCare Research Group (2023-GRIN-34459), Facultad de Enfermería de Cuenca, Universidad de Castilla-La Mancha, Cuenca, Spain; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile.
| | - Alicia Saz-Lara
- CarVasCare Research Group (2023-GRIN-34459), Facultad de Enfermería de Cuenca, Universidad de Castilla-La Mancha, Cuenca, Spain
| | - Irene Martínez-García
- CarVasCare Research Group (2023-GRIN-34459), Facultad de Enfermería de Cuenca, Universidad de Castilla-La Mancha, Cuenca, Spain
| | | | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
2
|
Wu Y, Wei Q, Lin J, Shang H, Ou R. Cognitive impairment, neuroimaging abnormalities, and their correlations in myotonic dystrophy: a comprehensive review. Front Cell Neurosci 2024; 18:1369332. [PMID: 38638300 PMCID: PMC11024338 DOI: 10.3389/fncel.2024.1369332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Myotonic dystrophy (DM) encompasses a spectrum of neuromuscular diseases characterized by myotonia, muscle weakness, and wasting. Recent research has led to the recognition of DM as a neurological disorder. Cognitive impairment is a central nervous system condition that has been observed in various forms of DM. Neuroimaging studies have increasingly linked DM to alterations in white matter (WM) integrity and highlighted the relationship between cognitive impairment and abnormalities in WM structure. This review aims to summarize investigations into cognitive impairment and brain abnormalities in individuals with DM and to elucidate the correlation between these factors and the potential underlying mechanisms contributing to these abnormalities.
Collapse
Affiliation(s)
| | | | | | | | - Ruwei Ou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Banerjee M, Venkatesh P, Azad SV. Ultra-widefield imaging and peripheral optical coherence tomography of peripheral reticular pigmentary degeneration (PRPD) in myotonic dystrophy. BMJ Case Rep 2023; 16:e258173. [PMID: 38035686 PMCID: PMC10689402 DOI: 10.1136/bcr-2023-258173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Affiliation(s)
- Mousumi Banerjee
- Dr. R.P. Center for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Pradeep Venkatesh
- Dr. R.P. Center for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Shorya Vardhan Azad
- Dr. R.P. Center for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, Delhi, India
| |
Collapse
|
4
|
Madrid DA, Knapp RA, Lynch D, Clemens P, Weaver AA, Puwanant A. Associations between lower extremity muscle fat fraction and motor performance in myotonic dystrophy type 2: A pilot study. Muscle Nerve 2023; 67:506-514. [PMID: 36938823 PMCID: PMC10898809 DOI: 10.1002/mus.27821] [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: 03/24/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/21/2023]
Abstract
INTRODUCTION/AIMS Although muscle structure measures from magnetic resonance imaging (MRI) have been used to assess disease severity in muscular dystrophies, little is known about how these measures are affected in myotonic dystrophy type 2 (DM2). We aim to characterize lower extremity muscle fat fraction (MFF) as a potential biomarker of disease severity, and evaluate its relationship with motor performance in DM2. METHODS 3-Tesla MRIs were obtained from nine patients with DM2 and six controls using a T1W-Dixon protocol. To calculate MFF, muscle volumes were segmented from proximal, middle, and distal regions of the thigh and calf. Associations between MFF and motor performance were calculated using Spearman's correlations (ρ). RESULTS Mean age of DM2 participants was 62 ± 11 y (89% female), and mean symptom duration was 20 ± 12 y. Compared to controls, the DM2 group had significantly higher MFF in the thigh and the calf segments (p-value = .002). The highest MFF at the thigh in DM2 was located in the posterior compartment (39.7 ± 12.9%) and at the calf was the lateral compartment (31.5 ± 8.7%). In the DM2 group, we found a strong correlation between the posterior thigh MFF and the 6-min walk test (ρ = -.90, p-value = .001). The lateral calf MFF was also strongly correlated with the step test (ρ = -0.82, p-value = .006). DISCUSSION Our pilot data suggest a potential correlation between lower extremity MFF and some motor performance tests in DM2. Longitudinal studies with larger sample sizes are required to validate MFF as a marker of disease severity in DM2.
Collapse
Affiliation(s)
- Diana A Madrid
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Rebecca A Knapp
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27101, USA
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina, 27109, USA
| | - Delanie Lynch
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Paula Clemens
- Department of Neurology, University of Pittsburgh School of Medicine and Department of Veterans Affairs Medical Center, Pittsburgh, Pennsylvania, 15213, USA
| | - Ashley A Weaver
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Araya Puwanant
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, 27157, USA
| |
Collapse
|
5
|
Gonzalez-Perez P, D'Ambrosio ES, Picher-Martel V, Chuang K, David WS, Amato AA. Parent-of-Origin Effect on the Age at Symptom Onset in Myotonic Dystrophy Type 2. Neurol Genet 2023; 9:e200073. [PMID: 37123986 PMCID: PMC10136683 DOI: 10.1212/nxg.0000000000200073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/02/2023] [Indexed: 05/02/2023]
Abstract
Background and Objectives The existence of clinical anticipation, congenital form, and parent-of-origin effect in myotonic dystrophy type 2 (DM2) remains uncertain. Here, we aimed at investigating whether there is a parent-of-origin effect on the age at the first DM2-related clinical manifestation. Methods We identified patients with genetically confirmed DM2 with known parental inheritance from (1) the electronic medical records of our institutions and (2) a systematic review of the literature following the PRISMA 2020 guidelines and recorded their age at and type of first disease-related symptom. We also interrogated the Myotonic Dystrophy Foundation Family Registry (MDFFR) for patients with DM2 who completed a survey including questions about parental inheritance and age at the first medical problem which they related to their DM2 diagnosis. Results A total of 26 patients with DM2 from 18 families were identified at our institutions as having maternal (n = 14) or paternal (n = 12) inheritance of the disease, whereas our systematic review of the literature rendered a total of 61 patients with DM2 from 41 families reported by 24 eligible articles as having maternal (n = 40) or paternal (n = 21) inheritance of the disease. Both cohorts were combined for downstream analyses. Up to 61% and 58% of patients had muscle-related symptoms as the first disease manifestation in maternally and paternally inherited DM2 subgroups, respectively. Four patients developed hypotonia at birth and/or delayed motor milestones early in life, and 7 had nonmuscular presentations (2 had cardiac events within the second decade of life and 5 had cataracts), all of them with maternal inheritance. A maternal inheritance was associated with an earlier (within the first 3 decades of life) age at symptom onset relative to a paternal inheritance in this combined cohort, and this association was independent of the patient's sex (OR [95% CI] = 4.245 [1.429-13.820], p = 0.0117). However, this association was not observed in the MDFFR DM2 cohort (n = 127), possibly because age at onset was self-reported, and the information about the type of first symptom or medical problem that patients related to DM2 was lacking. Discussion A maternal inheritance may increase the risk of an early DM2 onset and of cataracts and cardiovascular events as first DM2 manifestations.
Collapse
Affiliation(s)
- Paloma Gonzalez-Perez
- Department of Neurology (P.G.-P., V.P.-M., K.C., W.S.D.), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Neurology (E.S.D.A.), Nationwide Children's Hospital, Columbus, OH; and Department of Neurology (V.P.-M., A.A.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - Eleonora S D'Ambrosio
- Department of Neurology (P.G.-P., V.P.-M., K.C., W.S.D.), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Neurology (E.S.D.A.), Nationwide Children's Hospital, Columbus, OH; and Department of Neurology (V.P.-M., A.A.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - Vincent Picher-Martel
- Department of Neurology (P.G.-P., V.P.-M., K.C., W.S.D.), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Neurology (E.S.D.A.), Nationwide Children's Hospital, Columbus, OH; and Department of Neurology (V.P.-M., A.A.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - Kathy Chuang
- Department of Neurology (P.G.-P., V.P.-M., K.C., W.S.D.), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Neurology (E.S.D.A.), Nationwide Children's Hospital, Columbus, OH; and Department of Neurology (V.P.-M., A.A.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - William S David
- Department of Neurology (P.G.-P., V.P.-M., K.C., W.S.D.), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Neurology (E.S.D.A.), Nationwide Children's Hospital, Columbus, OH; and Department of Neurology (V.P.-M., A.A.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| | - Anthony A Amato
- Department of Neurology (P.G.-P., V.P.-M., K.C., W.S.D.), Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Neurology (E.S.D.A.), Nationwide Children's Hospital, Columbus, OH; and Department of Neurology (V.P.-M., A.A.A.), Brigham Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Flemming BC, Savioli KA, Borruso LA, Perkins JN, Curtis J. Respiratory Failure in an Infant with Known Congenital Anomalies and Novel Genetic Defect. Neoreviews 2023; 24:e107-e111. [PMID: 36720688 DOI: 10.1542/neo.24-2-e107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Brittany C Flemming
- Department of Pediatrics; Department of Otolaryngology-Head and Neck Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Katrina A Savioli
- Department of Pediatrics; Department of Otolaryngology-Head and Neck Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Laura A Borruso
- Department of Pediatrics; Department of Otolaryngology-Head and Neck Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Jonathan N Perkins
- Department of Pediatrics; Department of Otolaryngology-Head and Neck Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| | - Jerri Curtis
- Department of Pediatrics; Department of Otolaryngology-Head and Neck Surgery, Walter Reed National Military Medical Center, Bethesda, MD
| |
Collapse
|
7
|
Papadimas GK, Papadopoulos C, Kekou K, Kartanou C, Kladi A, Nitsa E, Sofocleous C, Tsanou E, Sarmas I, Kaninia S, Chroni E, Tsivgoulis G, Kimiskidis V, Arnaoutoglou M, Stefanis L, Panas M, Koutsis G, Karadima G, Traeger-Synodinos J. A Greek National Cross-Sectional Study on Myotonic Dystrophies. Int J Mol Sci 2022; 23:ijms232415507. [PMID: 36555146 PMCID: PMC9778724 DOI: 10.3390/ijms232415507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Myotonic Dystrophies (DM, Dystrophia Myotonia) are autosomal dominant inherited myopathies with a high prevalence across different ethnic regions. Despite some differences, mainly due to the pattern of muscle involvement and the age of onset, both forms, DM1 and DM2, share many clinical and genetic similarities. In this study, we retrospectively analyzed the medical record files of 561 Greek patients, 434 with DM1 and 127 with DM2 diagnosed in two large academic centers between 1994-2020. The mean age at onset of symptoms was 26.2 ± 15.3 years in DM1 versus 44.4 ± 17.0 years in DM2 patients, while the delay of diagnosis was 10 and 7 years for DM1 and DM2 patients, respectively. Muscle weakness was the first symptom in both types, while myotonia was more frequent in DM1 patients. Multisystemic involvement was detected in the great majority of patients, with cataracts being one of the most common extramuscular manifestations, even in the early stages of disease expression. In conclusion, the present work, despite some limitations arising from the retrospective collection of data, is the first record of a large number of Greek patients with myotonic dystrophy and emphasizes the need for specialized neuromuscular centers that can provide genetic counseling and a multidisciplinary approach.
Collapse
Affiliation(s)
- Georgios K. Papadimas
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- Correspondence: or ; Tel.: +30-210-7289152; Fax: +30-210-7216474
| | - Constantinos Papadopoulos
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Kyriaki Kekou
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, “Ag. Sofia” Children’s Hospital, 11527 Athens, Greece
| | - Chrisoula Kartanou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Athina Kladi
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Evangelia Nitsa
- Postgraduate Program in Biostatistics School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christalena Sofocleous
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, “Ag. Sofia” Children’s Hospital, 11527 Athens, Greece
| | - Evangelia Tsanou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Ioannis Sarmas
- Department of Neurology, University Hospital of Ioannina, University of Ioannina, 45500 Ioannina, Greece
| | - Stefania Kaninia
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Elisabeth Chroni
- Department of Neurology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, “Attikon” University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Vasilios Kimiskidis
- 1st Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Marianthi Arnaoutoglou
- Department of Clinical Neurophysiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Leonidas Stefanis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Marios Panas
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Georgios Koutsis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Georgia Karadima
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Joanne Traeger-Synodinos
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, “Ag. Sofia” Children’s Hospital, 11527 Athens, Greece
| |
Collapse
|
8
|
Soltanzadeh P. Myotonic Dystrophies: A Genetic Overview. Genes (Basel) 2022; 13:367. [PMID: 35205411 PMCID: PMC8872148 DOI: 10.3390/genes13020367] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophies (DM) are the most common muscular dystrophies in adults, which can affect other non-skeletal muscle organs such as the heart, brain and gastrointestinal system. There are two genetically distinct types of myotonic dystrophy: myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2), both dominantly inherited with significant overlap in clinical manifestations. DM1 results from CTG repeat expansions in the 3'-untranslated region (3'UTR) of the DMPK (dystrophia myotonica protein kinase) gene on chromosome 19, while DM2 is caused by CCTG repeat expansions in intron 1 of the CNBP (cellular nucleic acid-binding protein) gene on chromosome 3. Recent advances in genetics and molecular biology, especially in the field of RNA biology, have allowed better understanding of the potential pathomechanisms involved in DM. In this review article, core clinical features and genetics of DM are presented followed by a discussion on the current postulated pathomechanisms and therapeutic approaches used in DM, including the ones currently in human clinical trial phase.
Collapse
Affiliation(s)
- Payam Soltanzadeh
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
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]
|
10
|
Peric S, Rakocevic-Stojanovic V, Meola G. Cerebral involvement and related aspects in myotonic dystrophy type 2. Neuromuscul Disord 2021; 31:681-694. [PMID: 34244019 DOI: 10.1016/j.nmd.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/18/2023]
Abstract
Myotonic dystrophy type 2 (DM2) is an autosomal dominant multisystemic disorder caused by CCTG repeats expansion in the first intron of the CNBP gene. In this review we focus on the brain involvement in DM2, including its pathogenic mechanisms, microstructural, macrostructural and functional brain changes, as well as the effects of all these impairments on patients' everyday life. We also try to understand how brain abnormalities in DM2 should be adequately measured and potentially treated. The most important pathogenetic mechanisms in DM2 are RNA gain-of-function and repeat-associated non-ATG (RAN) translation. One of the main neuroimaging findings in DM2 is the presence of diffuse periventricular white matter hyperintensity lesions (WMHLs). Brain atrophy has been described in DM2 patients, but it is not clear if it is mostly caused by a decrease of the white or gray matter volume. The most commonly reported specific cognitive symptoms in DM2 are dysexecutive syndrome, visuospatial and memory impairments. Fatigue, sleep-related disorders and pain are also frequent in DM2. The majority of key symptoms and signs in DM2 has a great influence on patients' daily lives, their psychological status, economic situation and quality of life.
Collapse
Affiliation(s)
- Stojan Peric
- Neurology Clinic, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Giovanni Meola
- Department of Neurorehabilitation Sciences, Casa Di Cura del Policlinico, Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Meola G. Myotonic dystrophy type 2: the 2020 update. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2020; 39:222-234. [PMID: 33458578 PMCID: PMC7783423 DOI: 10.36185/2532-1900-026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/26/2022]
Abstract
The myotonic dystrophies are the commonest cause of adult-onset muscular dystrophy. Phenotypes of DM1 and DM2 are similar, but there are some important differences, including the presence or absence of congenital form, muscles primarily affected (distal vs proximal), involved muscle fiber types (type 1 vs type 2 fibers), and some associated multisystemic phenotypes. There is currently no cure for the myotonic dystrophies but effective management significantly reduces the morbidity and mortality of patients. For the enormous understanding of the molecular pathogenesis of myotonic dystrophy type 1 and myotonic dystrophy type 2, these diseases are now called "spliceopathies" and are mediated by a primary disorder of RNA rather than proteins. Despite clinical and genetic similarities, myotonic dystrophy type 1 and type 2 are distinct disorders requiring different diagnostic and management strategies. Gene therapy for myotonic dystrophy type 1 and myotonic dystrophy type 2 appears to be very close and the near future is an exciting time for clinicians and patients.
Collapse
Affiliation(s)
- Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, Italy.,Department of Neurorehabilitation Sciences, Casa di Cura del Policlinico, Milan, Italy
| |
Collapse
|
12
|
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: 1.3] [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
|
13
|
Taylor K, Sobczak K. Intrinsic Regulatory Role of RNA Structural Arrangement in Alternative Splicing Control. Int J Mol Sci 2020; 21:ijms21145161. [PMID: 32708277 PMCID: PMC7404189 DOI: 10.3390/ijms21145161] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Alternative splicing is a highly sophisticated process, playing a significant role in posttranscriptional gene expression and underlying the diversity and complexity of organisms. Its regulation is multilayered, including an intrinsic role of RNA structural arrangement which undergoes time- and tissue-specific alterations. In this review, we describe the principles of RNA structural arrangement and briefly decipher its cis- and trans-acting cellular modulators which serve as crucial determinants of biological functionality of the RNA structure. Subsequently, we engage in a discussion about the RNA structure-mediated mechanisms of alternative splicing regulation. On one hand, the impairment of formation of optimal RNA structures may have critical consequences for the splicing outcome and further contribute to understanding the pathomechanism of severe disorders. On the other hand, the structural aspects of RNA became significant features taken into consideration in the endeavor of finding potential therapeutic treatments. Both aspects have been addressed by us emphasizing the importance of ongoing studies in both fields.
Collapse
|
14
|
Nicolau S, Liewluck T, Milone M. Myopathies with finger flexor weakness: Not only inclusion-body myositis. Muscle Nerve 2020; 62:445-454. [PMID: 32478919 DOI: 10.1002/mus.26914] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022]
Abstract
Muscle disorders are characterized by differential involvement of various muscle groups. Among these, weakness predominantly affecting finger flexors is an uncommon pattern, most frequently found in sporadic inclusion-body myositis. This finding is particularly significant when the full range of histopathological findings of inclusion-body myositis is not found on muscle biopsy. Prominent finger flexor weakness, however, is also observed in other myopathies. It occurs commonly in myotonic dystrophy types 1 and 2. In addition, individual reports and small case series have documented finger flexor weakness in sarcoid and amyloid myopathy, and in inherited myopathies caused by ACTA1, CRYAB, DMD, DYSF, FLNC, GAA, GNE, HNRNPDL, LAMA2, MYH7, and VCP mutations. Therefore, the finding of finger flexor weakness requires consideration of clinical, myopathological, genetic, electrodiagnostic, and sometimes muscle imaging findings to establish a diagnosis.
Collapse
Affiliation(s)
- Stefan Nicolau
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| | - Teerin Liewluck
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| | - Margherita Milone
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| |
Collapse
|
15
|
La Rosa P, Petrillo S, Bertini ES, Piemonte F. Oxidative Stress in DNA Repeat Expansion Disorders: A Focus on NRF2 Signaling Involvement. Biomolecules 2020; 10:biom10050702. [PMID: 32369911 PMCID: PMC7277112 DOI: 10.3390/biom10050702] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or the gain of function of its transcribed mRNA or translated product represent the major pathogenic effect of these pathologies, mitochondrial dysfunction and imbalance in redox homeostasis are reported as common features in these disorders, deeply affecting their severity and progression. In this review, we examine the role that the redox imbalance plays in the pathological mechanisms of DNA expansion disorders and the recent advances on antioxidant treatments, particularly focusing on the expression and the activity of the transcription factor NRF2, the main cellular regulator of the antioxidant response.
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW This article describes the clinical features, pathogenesis, prevalence, diagnosis, and management of myotonic dystrophy type 1 and myotonic dystrophy type 2. RECENT FINDINGS The prevalence of myotonic dystrophy type 1 is better understood than the prevalence of myotonic dystrophy type 2, and new evidence indicates that the risk of cancer is increased in patients with the myotonic dystrophies. In addition, descriptions of the clinical symptoms and relative risks of comorbidities such as cardiac arrhythmias associated with myotonic dystrophy type 1 have been improved. SUMMARY Myotonic dystrophy type 1 and myotonic dystrophy type 2 are both characterized by progressive muscle weakness, early-onset cataracts, and myotonia. However, both disorders have multisystem manifestations that require a comprehensive management plan. While no disease-modifying therapies have yet been identified, advances in therapeutic development have a promising future.
Collapse
|
17
|
Gois Beghini D, Iwao Horita S, Monteiro da Fonseca Cardoso L, Anastacio Alves L, Nagaraju K, Henriques-Pons A. A Promising Future for Stem-Cell-Based Therapies in Muscular Dystrophies-In Vitro and In Vivo Treatments to Boost Cellular Engraftment. Int J Mol Sci 2019; 20:ijms20215433. [PMID: 31683627 PMCID: PMC6861917 DOI: 10.3390/ijms20215433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/28/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophies (MD) are a group of genetic diseases that lead to skeletal muscle wasting and may affect many organs (multisystem). Unfortunately, no curative therapies are available at present for MD patients, and current treatments mainly address the symptoms. Thus, stem-cell-based therapies may present hope for improvement of life quality and expectancy. Different stem cell types lead to skeletal muscle regeneration and they have potential to be used for cellular therapies, although with several limitations. In this review, we propose a combination of genetic, biochemical, and cell culture treatments to correct pathogenic genetic alterations and to increase proliferation, dispersion, fusion, and differentiation into new or hybrid myotubes. These boosted stem cells can also be injected into pretreate recipient muscles to improve engraftment. We believe that this combination of treatments targeting the limitations of stem-cell-based therapies may result in safer and more efficient therapies for MD patients. Matricryptins have also discussed.
Collapse
Affiliation(s)
- Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Samuel Iwao Horita
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | | | - Luiz Anastacio Alves
- Laboratório de Comunicação Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, NY 13902, USA.
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| |
Collapse
|
18
|
Higgs C, Hilbert JE, Wood L, Martens WB, Marini-Bettolo C, Nikolenko N, Alsaggaf R, Lochmüller H, Moxley RT, Greene MH, Wang Y, Gadalla SM. Reproductive Cancer Risk Factors in Women With Myotonic Dystrophy (DM): Survey Data From the US and UK DM Registries. Front Neurol 2019; 10:1071. [PMID: 31681146 PMCID: PMC6797599 DOI: 10.3389/fneur.2019.01071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/23/2019] [Indexed: 11/24/2022] Open
Abstract
Introduction: Recent evidence demonstrates that women with myotonic dystrophy type 1 are at increased risk of reproductive organ tumors. However, studies of reproductive cancer risk factors in those patients are lacking. Methods: Using questionnaires, we collected and analyzed personal history information related to cancer risk factors from women enrolled in a UK and US registry for myotonic dystrophy (dystrophia myotonica; DM) patients. Results: The survey was completed by 242 DM type 1 (DM1) and 44 DM type 2 (DM2) women enrolled in the UK Registry (N = 124) and the US National Registry (N = 162). The mean age at DM1 diagnosis was 33.8 years (standard deviation, SD = 13.2) and for DM2 was 49.2 (SD = 13.0). Mean age at survey was 48.7 (SD = 12.8) and 59.1 years (SD = 12.8) for DM1 and DM2, respectively. There were no statistically significant differences between DM1 and DM2 regarding menstrual history or fertility-related factors. Yet, women with DM2 were more likely to have used menopausal hormone therapy (HT) than women with DM1 (52.3 vs. 22.1%, p < 0.0001), and more women with DM2 had a hysterectomy (53.5 vs. 29.5%, p < 0.01). These differences were not statistically significant after age adjustment (OR = 2.00, p = 0.08, and OR = 1.40, p = 0.38, respectively). The frequency of self-reported reproductive organ tumors was not significantly different comparing DM1 to DM2 (p = 0.28). However, the data suggested that women with DM2 appear to have a lower risk of malignant tumors compared to those with DM1 (OR = 0.72, p = 0.69). Discussion: Our study is the first to characterize a wide range of reproductive risk factors in women with DM. We observed no significant differences between DM1 and DM2 in the factors that were evaluated, which suggests that the known excesses of ovarian and endometrial cancer previously reported in women with DM1 cannot be attributed to greater prevalence of standard cancer-related reproductive risk factors. Larger studies evaluating the possible link between reproductive cancer risk factors and risk of tumors in women with DM are needed.
Collapse
Affiliation(s)
- Cecilia Higgs
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, United States
| | - James E Hilbert
- Department of Neurology, Neuromuscular Disease Center, University of Rochester Medical Center, Rochester, NY, United States
| | - Libby Wood
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - William B Martens
- Department of Neurology, Neuromuscular Disease Center, University of Rochester Medical Center, Rochester, NY, United States
| | - Chiara Marini-Bettolo
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nikoletta Nikolenko
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Rotana Alsaggaf
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, United States
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation Barcelona, Institute of Science and Technology (BIST), Barcelona, Spain.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Richard T Moxley
- Department of Neurology, Neuromuscular Disease Center, University of Rochester Medical Center, Rochester, NY, United States
| | - Mark H Greene
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, United States
| | - Youjin Wang
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, United States
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
19
|
Taylor K, Sznajder LJ, Cywoniuk P, Thomas JD, Swanson MS, Sobczak K. MBNL splicing activity depends on RNA binding site structural context. Nucleic Acids Res 2019; 46:9119-9133. [PMID: 29955876 PMCID: PMC6158504 DOI: 10.1093/nar/gky565] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/16/2018] [Indexed: 02/02/2023] Open
Abstract
Muscleblind-like (MBNL) proteins are conserved RNA-binding factors involved in alternative splicing (AS) regulation during development. While AS is controlled by distribution of MBNL paralogs and isoforms, the affinity of these proteins for specific RNA-binding regions and their location within transcripts, it is currently unclear how RNA structure impacts MBNL-mediated AS regulation. Here, we defined the RNA structural determinants affecting MBNL-dependent AS activity using both cellular and biochemical assays. While enhanced inclusion of MBNL-regulated alternative exons is controlled by the arrangement and number of MBNL binding sites within unstructured RNA, when these sites are embedded in a RNA hairpin MBNL binds preferentially to one side of stem region. Surprisingly, binding of MBNL proteins to RNA targets did not entirely correlate with AS efficiency. Moreover, comparison of MBNL proteins revealed structure-dependent competitive behavior between the paralogs. Our results showed that the structure of targeted RNAs is a prevalent component of the mechanism of alternative splicing regulation by MBNLs.
Collapse
Affiliation(s)
- Katarzyna Taylor
- Laboratory of Gene Therapy, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Lukasz J Sznajder
- Center for NeuroGenetics and the Genetics Institute, Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, 2033 Mowry Road, Gainesville, FL 32610, USA
| | - Piotr Cywoniuk
- Laboratory of Gene Therapy, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - James D Thomas
- Center for NeuroGenetics and the Genetics Institute, Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, 2033 Mowry Road, Gainesville, FL 32610, USA.,Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Maurice S Swanson
- Center for NeuroGenetics and the Genetics Institute, Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, 2033 Mowry Road, Gainesville, FL 32610, USA
| | - Krzysztof Sobczak
- Laboratory of Gene Therapy, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| |
Collapse
|
20
|
Hale MA, Johnson NE, Berglund JA. Repeat-associated RNA structure and aberrant splicing. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:194405. [PMID: 31323433 DOI: 10.1016/j.bbagrm.2019.07.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022]
Abstract
Over 30 hereditary disorders attributed to the expansion of microsatellite repeats have been identified. Despite variant nucleotide content, number of consecutive repeats, and different locations in the genome, many of these diseases have pathogenic RNA gain-of-function mechanisms. The repeat-containing RNAs can form structures in vitro predicted to contribute to the disease through assembly of intracellular RNA aggregates termed foci. The expanded repeat RNAs within these foci sequester RNA binding proteins (RBPs) with important roles in the regulation of RNA metabolism, most notably alternative splicing (AS). These deleterious interactions lead to downstream alterations in transcriptome-wide AS directly linked with disease symptoms. This review summarizes existing knowledge about the association between the repeat RNA structures and RBPs as well as the resulting aberrant AS patterns, specifically in the context of myotonic dystrophy. The connection between toxic, structured RNAs and dysregulation of AS in other repeat expansion diseases is also discussed. This article is part of a Special Issue entitled: RNA structure and splicing regulation edited by Francisco Baralle, Ravindra Singh and Stefan Stamm.
Collapse
Affiliation(s)
- Melissa A Hale
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Nicholas E Johnson
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - J Andrew Berglund
- The RNA Institute, Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA.
| |
Collapse
|
21
|
Wahbi K, Furling D. Cardiovascular manifestations of myotonic dystrophy. Trends Cardiovasc Med 2019; 30:232-238. [PMID: 31213350 DOI: 10.1016/j.tcm.2019.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 11/25/2022]
Abstract
Patients with myotonic dystrophy, the most common neuromuscular dystrophy in adults, have a high prevalence of arrhythmic complications with increased cardiovascular mortality and high risk for sudden death. Sudden death prevention is central and relies on annual follow-up and prophylactic permanent pacing in patients with conduction defects on electrocardiogram and/or infrahisian blocks on electrophysiological study. Implantable cardiac defibrillator therapy may be indicated in patients with ventricular tachyarrhythmia.
Collapse
Affiliation(s)
- Karim Wahbi
- APHP, Cochin Hospital, Cardiology Department, Centre de Référence de Pathologie Neuromusculaire, Nord Est, Ile de France, Paris-Descartes, Sorbonne Paris Cité University, Cochin Hospital, 27 Rue du Faubourg Saint Jacques, 75679 Paris Cedex 14 Paris, France.
| | - Denis Furling
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
22
|
De Antonio M, Dogan C, Daidj F, Eymard B, Puymirat J, Mathieu J, Gagnon C, Katsahian S, Hamroun D, Bassez G. The DM-scope registry: a rare disease innovative framework bridging the gap between research and medical care. Orphanet J Rare Dis 2019; 14:122. [PMID: 31159885 PMCID: PMC6547518 DOI: 10.1186/s13023-019-1088-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/06/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The relevance of registries as a key component for developing clinical research for rare diseases (RD) and improving patient care has been acknowledged by most stakeholders. As recent studies pointed to several limitations of RD registries our challenge was (1) to improve standardization and data comparability; (2) to facilitate interoperability between existing RD registries; (3) to limit the amount of incomplete data; (4) to improve data quality. This report describes the innovative concept of the DM-Scope Registry that was developed to achieve these objectives for Myotonic Dystrophy (DM), a prototypical example of highly heterogeneous RD. By the setting up of an integrated platform attractive for practitioners use, we aimed to promote DM epidemiology, clinical research and patients care management simultaneously. RESULTS The DM-Scope Registry is a result of the collaboration within the French excellence network established by the National plan for RDs. Inclusion criteria is all genetically confirmed DM individuals, independently of disease age of onset. The dataset includes social-demographic data, clinical features, genotype, and biomaterial data, and is adjustable for clinical trial data collection. To date, the registry has a nationwide coverage, composed of 55 neuromuscular centres, encompassing the whole disease clinical and genetic spectrum. This widely used platform gathers almost 3000 DM patients (DM1 n = 2828, DM2 n = 142), both children (n = 322) and adults (n = 2648), which accounts for > 20% of overall registered DM patients internationally. The registry supported 10 research studies of various type i.e. observational, basic science studies and patient recruitment for clinical trials. CONCLUSION The DM-Scope registry represents the largest collection of standardized data for the DM population. Our concept improved collaboration among health care professionals by providing annual follow-up of quality longitudinal data collection. The combination of clinical features and biomolecular materials provides a comprehensive view of the disease in a given population. DM-Scope registry proves to be a powerful device for promoting both research and medical care that is suitable to other countries. In the context of emerging therapies, such integrated platform contributes to the standardisation of international DM research and for the design of multicentre clinical trials. Finally, this valuable model is applicable to other RDs.
Collapse
Affiliation(s)
- Marie De Antonio
- Neuromuscular Reference Center, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne University, Paris Descartes University, Paris, France
| | - Céline Dogan
- Neuromuscular Reference Center, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Ferroudja Daidj
- Neuromuscular Reference Center, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Bruno Eymard
- Neuromuscular Reference Center, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | | | - Jean Mathieu
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires (GRIMN), CIUSSS du Saguenay-Lac-St-Jean, Québec, Canada
| | - Cynthia Gagnon
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires (GRIMN), CIUSSS du Saguenay-Lac-St-Jean, Québec, Canada
- Centre de recherche Charles-Le-Moyne-Saguenay-Lac-St-Jean sur les innovations en santé (CR-CSIS), Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Québec, Canada
| | - Sandrine Katsahian
- INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne University, Paris Descartes University, Paris, France
- Unit of Epidemiology and Clinical Research, AP-HP, Georges-Pompidou Hospital, Paris, France
| | - Dalil Hamroun
- University Institute of Clinical Research, CHU, Montpellier, France
| | - Guillaume Bassez
- Neuromuscular Reference Center, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, Research Center in Myology, Sorbonne University, Paris, France
| |
Collapse
|
23
|
Kim EY, Barefield DY, Vo AH, Gacita AM, Schuster EJ, Wyatt EJ, Davis JL, Dong B, Sun C, Page P, Dellefave-Castillo L, Demonbreun A, Zhang HF, McNally EM. Distinct pathological signatures in human cellular models of myotonic dystrophy subtypes. JCI Insight 2019; 4:122686. [PMID: 30730308 DOI: 10.1172/jci.insight.122686] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 01/31/2019] [Indexed: 01/06/2023] Open
Abstract
Myotonic dystrophy (DM) is the most common autosomal dominant muscular dystrophy and encompasses both skeletal muscle and cardiac complications. DM is nucleotide repeat expansion disorder in which type 1 (DM1) is due to a trinucleotide repeat expansion on chromosome 19 and type 2 (DM2) arises from a tetranucleotide repeat expansion on chromosome 3. Developing representative models of DM in animals has been challenging due to instability of nucleotide repeat expansions, especially for DM2, which is characterized by nucleotide repeat expansions often greater than 5,000 copies. To investigate mechanisms of human DM, we generated cellular models of DM1 and DM2. We used regulated MyoD expression to reprogram urine-derived cells into myotubes. In this myogenic cell model, we found impaired dystrophin expression, in the presence of muscleblind-like 1 (MBNL1) foci, and aberrant splicing in DM1 but not in DM2 cells. We generated induced pluripotent stem cells (iPSC) from healthy controls and DM1 and DM2 subjects, and we differentiated these into cardiomyocytes. DM1 and DM2 cells displayed an increase in RNA foci concomitant with cellular differentiation. iPSC-derived cardiomyocytes from DM1 but not DM2 had aberrant splicing of known target genes and MBNL sequestration. High-resolution imaging revealed tight association between MBNL clusters and RNA foci in DM1. Ca2+ transients differed between DM1- and DM2 iPSC-derived cardiomyocytes, and each differed from healthy control cells. RNA-sequencing from DM1- and DM2 iPSC-derived cardiomyocytes revealed distinct misregulation of gene expression, as well as differential aberrant splicing patterns. Together, these data support that DM1 and DM2, despite some shared clinical and molecular features, have distinct pathological signatures.
Collapse
Affiliation(s)
- Ellis Y Kim
- Molecular Pathogenesis and Molecular Medicine, The University of Chicago, Chicago, Illinois, USA
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andy H Vo
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, Illinois, USA
| | - Anthony M Gacita
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Emma J Schuster
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Eugene J Wyatt
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Biqin Dong
- Department of Biomedical Engineering and.,Department of Mechanical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Cheng Sun
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Patrick Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lisa Dellefave-Castillo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alexis Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
24
|
Romigi A, Franco V, Placidi F, Liguori C, Rastelli E, Vitrani G, Centonze D, Massa R. Comparative Sleep Disturbances in Myotonic Dystrophy Types 1 and 2. Curr Neurol Neurosci Rep 2018; 18:102. [DOI: 10.1007/s11910-018-0903-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Abstract
Abstract:Introduction:There are two case reports of patients who had proximal myotonic myopathy (PROMM) / myotonic dystrophy (DM) Type 1 and parkinsonism. The combination of myotonic myopathy and parkinsonism is so rare that it may appear to be just a coincidence. However, previous neuropathological examinations of patients who had myotonic dystrophy showed that there were intracytoplasmic inclusion bodies in the nigra and striatum, which raises the possibility that myotonic myopathy may be associated with parkinsonism. In this report we describe a patient with PROMM and a clinically definite parkinsonism to highlight this possibility.Case Report:A 65-year-old man developed proximal muscle weakness, myotonia and atrophy around the age of 55 and was diagnosed as having PROMM at the age of 62. Needle electromyography and muscle biopsy supported the diagnosis. A gene study of the DM Type 1 showed a normal CTG repeat length. At age 63, he developed rest tremor, bradykinesia, hypomimia, stooped posture, and gait disturbance. The postural instability worsened rapidly. The tremor and rigidity were much worse in his right side, where myotonia was more severe. Levodopa therapy was only partially effective.Conclusion:This is a case report of a patient with PROMM that shows an association with a rapidly progressive form of parkinsonism. We suggest that this may be a novel form of a neurodegenerative disorder, which we name ‘Parkinsonism- Myotonic Myopathy-Complex’.
Collapse
|
26
|
Minnerop M, Gliem C, Kornblum C. Current Progress in CNS Imaging of Myotonic Dystrophy. Front Neurol 2018; 9:646. [PMID: 30186217 PMCID: PMC6110944 DOI: 10.3389/fneur.2018.00646] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/18/2018] [Indexed: 01/18/2023] Open
Abstract
Neuroimaging in myotonic dystrophies provided a major contribution to the insight into brain involvement which is highly prevalent in these multisystemic disorders. Particular in Myotonic Dystrophy Type 1, conventional MRI first revealed hyperintense white matter lesions, predominantly localized in the anterior temporal lobe. Brain atrophy and ventricle enlargement were additional early findings already described almost 30 years ago. Since then, more advanced and sophisticated imaging methods have been applied in Myotonic Dystrophy Types 1 and 2. Involvement of actually normal appearing white matter and widespread cortical affection in PET studies were key results toward the recognition of diffuse and not only focally localized brain pathology in vivo. Later, structural abnormalities of both, gray and white matter, have been found in both forms of the disorder, albeit more prominent in myotonic dystrophy type 1. In Type 1, a consistent widespread cortical and subcortical involvement of gray and white matter affecting all lobes, brainstem and cerebellum was observed. Spectroscopy studies gave additional evidence of neuronal and glial damage in both types. Central questions regarding the origin and spatiotemporal evolution of the CNS involvement and its relevance for clinical symptoms had already been raised 30 years ago, however are still not answered. Results of correlation analyses between neuroimaging and clinical parameters are diverse and with few exceptions not well reproducible across studies. It may be related to the fact that most of the reported studies included only small numbers of subjects, sometimes even not separating Myotonic Dystrophy Type 1 from Type 2. But this heterogeneity may also support the current point of view that the clinical impairments are not simply linked to specific and regionally circumscribed structural or functional brain alterations. It seems more convincing that disturbed networks build the functional and structural substrate of clinical symptoms in these disorders as it is proposed in other neuropsychiatric diseases. Consecutively, structural and functional network analyses may provide additional information regarding the link between brain pathology and clinical symptoms. Up to now, only cross-sectional neuroimaging studies have been published. To analyze the temporal evolution of brain affection, longitudinal studies are urgently needed, and systematic natural history data would be useful to identify potential biomarkers for therapeutic studies.
Collapse
Affiliation(s)
- Martina Minnerop
- Institute of Neuroscience and Medicine (INM-1), Research Center Juelich, Juelich, Germany.,Department of Neurology and Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Center for Movement Disorders and Neuromodulation, Heinrich-Heine University, Düsseldorf, Germany
| | - Carla Gliem
- Department of Neurology, University Hospital of Bonn, Bonn, Germany
| | - Cornelia Kornblum
- Department of Neurology, University Hospital of Bonn, Bonn, Germany.,Center for Rare Diseases Bonn (ZSEB), University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
27
|
SCN4A as modifier gene in patients with myotonic dystrophy type 2. Sci Rep 2018; 8:11058. [PMID: 30038349 PMCID: PMC6056531 DOI: 10.1038/s41598-018-29302-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/02/2018] [Indexed: 01/05/2023] Open
Abstract
A patient with an early severe myotonia diagnosed for Myotonic Dystrophy type 2 (DM2) was found bearing the combined effects of DM2 mutation and Nav1.4 S906T substitution. To investigate the mechanism underlying his atypical phenotype,whole-cell patch-clamp in voltage- and current-clamp mode was performed in myoblasts and myotubes obtained from his muscle biopsy. Results characterizing the properties of the sodium current and of the action potentials have been compared to those obtained in muscle cells derived from his mother, also affected by DM2, but without the S906T polymorphism. A faster inactivation kinetics and a +5 mV shift in the availability curve were found in the sodium current recorded in patient’s myoblasts compared to his mother. 27% of his myotubes displayed spontaneous activity. Patient’s myotubes showing a stable resting membrane potential had a lower rheobase current respect to the mother’s while the overshoot and the maximum slope of the depolarizing phase of action potential were higher. These findings suggest that SCN4A polymorphisms may be responsible for a higher excitability of DM2 patients sarcolemma, supporting the severe myotonic phenotype observed. We suggest SCN4A as a modifier factor and that its screening should be performed in DM2 patients with uncommon clinical features.
Collapse
|
28
|
Chakraborty M, Llamusi B, Artero R. Modeling of Myotonic Dystrophy Cardiac Phenotypes in Drosophila. Front Neurol 2018; 9:473. [PMID: 30061855 PMCID: PMC6054993 DOI: 10.3389/fneur.2018.00473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/31/2018] [Indexed: 01/07/2023] Open
Abstract
After respiratory distress, cardiac dysfunction is the second most common cause of fatality associated with the myotonic dystrophy (DM) disease. Despite the prevalance of heart failure in DM, physiopathological studies on heart symptoms have been relatively scarce because few murine models faithfully reproduce the cardiac disease. Consequently, only a small number of candidate compounds have been evaluated in this specific phenotype. To help cover this gap Drosophila combines the amenability of its invertebrate genetics with the possibility of quickly acquiring physiological parameters suitable for meaningful comparisons with vertebrate animal models and humans. Here we review available descriptions of cardiac disease in DM type 1 and type 2, and three recent papers reporting the cardiac toxicity of non-coding CUG (DM1) and CCUG (DM2) repeat RNA in flies. Notably, flies expressing CUG or CCUG RNA in their hearts developed strong arrhythmias and had reduced fractional shortening, which correlates with similar phenotypes in DM patients. Overexpression of Muscleblind, which is abnormally sequestered by CUG and CCUG repeat RNA, managed to strongly suppress arrhythmias and fractional shortening, thus demonstrating that Muscleblind depletion causes cardiac phenotypes in flies. Importantly, small molecules pentamidine and daunorubicin were able to rescue cardiac phenotypes by releasing Muscleblind from sequestration. Taken together, fly heart models have the potential to make important contributions to the understanding of the molecular causes of cardiac dysfunction in DM and in the quick assessment of candidate therapeutics.
Collapse
Affiliation(s)
- Mouli Chakraborty
- Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain.,Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia, Spain.,CIPF-INCLIVA Joint Unit, Valencia, Spain
| | - Beatriz Llamusi
- Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain.,Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia, Spain.,CIPF-INCLIVA Joint Unit, Valencia, Spain
| | - Ruben Artero
- Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain.,Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia, Spain.,CIPF-INCLIVA Joint Unit, Valencia, Spain
| |
Collapse
|
29
|
Wurster CD, Ludolph AC. Antisense oligonucleotides in neurological disorders. Ther Adv Neurol Disord 2018; 11:1756286418776932. [PMID: 29854003 PMCID: PMC5971383 DOI: 10.1177/1756286418776932] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022] Open
Abstract
The introduction of genetics revolutionized the field of neurodegenerative and neuromuscular diseases and has provided considerable insight into the underlying pathomechanisms. Nevertheless, effective treatment options have been limited. This changed recently when antisense oligonucleotides (ASOs) could be translated from in vitro and experimental animal studies into clinical practice. In 2016, two ASOs were approved by the United States US Food and Drug Administration (FDA) and demonstrated remarkable efficacy in Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA). ASOs are synthetic single-stranded strings of nucleic acids. They selectively bind to specific premessenger ribonucleic acid (pre-mRNA)/mRNA sequences and alter protein synthesis by several mechanisms of action. Thus, apart from gene replacement, ASOs may therefore provide the most direct therapeutic strategy for influencing gene expression. In this review, we shall discuss basic mechanisms of ASO action, the role of chemical modifications needed to improve the pharmacodynamic and pharmacokinetic properties of ASOs, and we shall then focus on several ASOs developed for the treatment of neurodegenerative and neuromuscular disorders, including SMA, DMD, myotonic dystrophies, Huntington's disease, amyotrophic lateral sclerosis and Alzheimer's disease.
Collapse
Affiliation(s)
- Claudia D. Wurster
- Department of Neurology, University of Ulm, Oberer Eselsberg 45, Ulm, 89081, Germany
| | | |
Collapse
|
30
|
Thomas JD, Oliveira R, Sznajder ŁJ, Swanson MS. Myotonic Dystrophy and Developmental Regulation of RNA Processing. Compr Physiol 2018; 8:509-553. [PMID: 29687899 PMCID: PMC11323716 DOI: 10.1002/cphy.c170002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myotonic dystrophy (DM) is a multisystemic disorder caused by microsatellite expansion mutations in two unrelated genes leading to similar, yet distinct, diseases. DM disease presentation is highly variable and distinguished by differences in age-of-onset and symptom severity. In the most severe form, DM presents with congenital onset and profound developmental defects. At the molecular level, DM pathogenesis is characterized by a toxic RNA gain-of-function mechanism that involves the transcription of noncoding microsatellite expansions. These mutant RNAs disrupt key cellular pathways, including RNA processing, localization, and translation. In DM, these toxic RNA effects are predominantly mediated through the modulation of the muscleblind-like and CUGBP and ETR-3-like factor families of RNA binding proteins (RBPs). Dysfunction of these RBPs results in widespread RNA processing defects culminating in the expression of developmentally inappropriate protein isoforms in adult tissues. The tissue that is the focus of this review, skeletal muscle, is particularly sensitive to mutant RNA-responsive perturbations, as patients display a variety of developmental, structural, and functional defects in muscle. Here, we provide a comprehensive overview of DM1 and DM2 clinical presentation and pathology as well as the underlying cellular and molecular defects associated with DM disease onset and progression. Additionally, fundamental aspects of skeletal muscle development altered in DM are highlighted together with ongoing and potential therapeutic avenues to treat this muscular dystrophy. © 2018 American Physiological Society. Compr Physiol 8:509-553, 2018.
Collapse
Affiliation(s)
- James D. Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Ruan Oliveira
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Łukasz J. Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Maurice S. Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
31
|
Deregulation of RNA Metabolism in Microsatellite Expansion Diseases. ADVANCES IN NEUROBIOLOGY 2018; 20:213-238. [PMID: 29916021 DOI: 10.1007/978-3-319-89689-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RNA metabolism impacts different steps of mRNA life cycle including splicing, polyadenylation, nucleo-cytoplasmic export, translation, and decay. Growing evidence indicates that defects in any of these steps lead to devastating diseases in humans. This chapter reviews the various RNA metabolic mechanisms that are disrupted in Myotonic Dystrophy-a trinucleotide repeat expansion disease-due to dysregulation of RNA-Binding Proteins. We also compare Myotonic Dystrophy to other microsatellite expansion disorders and describe how some of these mechanisms commonly exert direct versus indirect effects toward disease pathologies.
Collapse
|
32
|
Hilbert JE, Barohn RJ, Clemens PR, Luebbe EA, Martens WB, McDermott MP, Parkhill AL, Tawil R, Thornton CA, Moxley RT. High frequency of gastrointestinal manifestations in myotonic dystrophy type 1 and type 2. Neurology 2017; 89:1348-1354. [PMID: 28855409 PMCID: PMC5649763 DOI: 10.1212/wnl.0000000000004420] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/07/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To analyze gastrointestinal (GI) manifestations, their progression over time, and medications being used to treat GI symptoms in a large cohort of patients with myotonic dystrophy types 1 (DM1) and 2 (DM2). METHODS We analyzed patient-reported data and medical records in a national registry cohort at baseline and 5 years. RESULTS At baseline, the majority of patients reported trouble swallowing in DM1 (55%; n = 499 of 913) and constipation in DM2 (53%; n = 96 of 180). Cholecystectomy occurred in 16.5% of patients with DM1 and 12.8% of patients with DM2, on average before 45 years of age. The use of medications indicated for gastroesophageal reflux disease was reported by 22.5% of DM1 and 18.9% of patients with DM2. Greater risk of a GI manifestation was associated with higher body mass index and longer disease duration in DM1 and female sex in DM2. At the 5-year follow-up, the most common new manifestations were trouble swallowing in patients with DM1 and constipation in patients with DM2. CONCLUSIONS GI manifestations were common in both DM1 and DM2, with a relatively high frequency of gallbladder removal in DM1 and DM2 occurring at a younger age compared to normative data in the literature. Studies are needed to determine the pathomechanism of how sex, weight gain, and duration of disease contribute to GI manifestations and how these manifestations affect quality of life and clinical care for patients with DM1 and DM2.
Collapse
Affiliation(s)
- James E Hilbert
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY.
| | - Richard J Barohn
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - Paula R Clemens
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - Elizabeth A Luebbe
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - William B Martens
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - Michael P McDermott
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - Amy L Parkhill
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - Rabi Tawil
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | - Charles A Thornton
- From the Departments of Neurology (J.E.H., E.A.L., W.B.M., M.P.M., R.T., C.A.T., R.T.M.) and Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; Department of Neurology (R.J.B.), University of Kansas Medical Center, Kansas City; Department of Neurology (P.R.C.), University of Pittsburgh and Department of Veterans Affairs Medical Center, PA; and Wegmans School of Pharmacy (A.L.P.), St. John Fisher College, Rochester, NY
| | | |
Collapse
|
33
|
Biomolecular diagnosis of myotonic dystrophy type 2: a challenging approach. J Neurol 2017; 264:1705-1714. [PMID: 28550479 DOI: 10.1007/s00415-017-8504-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/02/2017] [Indexed: 01/23/2023]
Abstract
Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are the most common adult form of muscular dystrophy, characterized by autosomal dominant progressive myopathy, myotonia, and multiorgan involvement. The onset and symptoms of the myotonic dystrophies are diverse, complicating their diagnoses and limiting a comprehensive approach to their clinical care. Diagnostic delay in DM2 is due not only to the heterogeneous phenotype and the aspecific onset but also to the unfamiliarity with the disorder by most clinicians. Moreover, the DM2 diagnostic odyssey is complicated by the difficulties to develop an accurate, robust, and cost-effective method for a routine molecular assay. The aim of this review is to underline by challenging approach the diagnostic limits and pitfalls that could results in failure to recognize the presence of DM2 disease. Understanding and preventing delays in DM2 diagnosis may facilitate family planning, improve symptom management in the short term, and facilitate more specific treatment in the long term.
Collapse
|
34
|
Gadalla SM, Hilbert JE, Martens WB, Givens S, Moxley RT, Greene MH. Pigmentation phenotype, photosensitivity and skin neoplasms in patients with myotonic dystrophy. Eur J Neurol 2017; 24:713-718. [PMID: 28317292 PMCID: PMC5464410 DOI: 10.1111/ene.13276] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/04/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND PURPOSE Recent studies have suggested a possible excess risk of skin neoplasms in patients with myotonic dystrophy (DM). Risk factors related to this observation have not been defined. METHOD Information regarding personal history of skin tumors, pigmentation phenotype, and skin reaction to sun exposure were collected from 266 DM patients who were enrolled in the US National Institutes of Health National Registry of Myotonic Dystrophy and Facioscapulohumeral Muscular Dystrophy Patients and Family Members. RESULTS Seventy-seven subjects reported having skin tumors that were either benign (n = 31), malignant (n = 32) or both (n = 14). Female gender [odds ratio (OR) = 2.27, 95% confidence interval (CI) 1.02-5.05, P = 0.04], older age (OR = 1.10, 95% CI 1.05-1.16, P < 0.001) and DM1 subtype (OR = 3.42, 95% CI 1.27-9.26, P = 0.02) were associated with a malignant skin tumor. The associations between malignant skin tumors and known risk factors [light eye color (OR = 1.62, 95% CI 0.78-3.39, P = 0.20), light skin complexion (OR = 1.31, 95% CI 0.63-2.73, P = 0.48) and moderate/extensive face freckles (OR = 1.47, 95% CI 0.50-4.34, P = 0.49)] were modest. Strong, but not statistically significant, associations were noted with sunburn reactions when exposed to sunlight (OR = 4.28, 95% CI 0.91-19.95, P = 0.06, and OR = 2.19, 95% CI 0.67-7.09, P = 0.19, for sunburn with and without blistering, respectively). CONCLUSIONS Although our study was limited by small sample size, the risk factors for malignant skin tumors in DM strongly resemble the general population. It is recommended that DM patients adhere to sun exposure protective behavior.
Collapse
Affiliation(s)
- Shahinaz M. Gadalla
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - James E. Hilbert
- Department of Neurology, Neuromuscular Disease Center, University of Rochester Medical Center, Rochester, NY, USA
| | - William B. Martens
- Department of Neurology, Neuromuscular Disease Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Shannon Givens
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Richard T. Moxley
- Department of Neurology, Neuromuscular Disease Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Mark H. Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
35
|
Meola G, Cardani R. Myotonic dystrophy type 2 and modifier genes: an update on clinical and pathomolecular aspects. Neurol Sci 2017; 38:535-546. [PMID: 28078562 DOI: 10.1007/s10072-016-2805-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022]
Abstract
Myotonic dystrophy (DM) is the most common adult muscular dystrophy, characterized by autosomal dominant progressive myopathy, myotonia, and multiorgan involvement. To date, two distinct forms caused by similar mutations in two different genes have been identified: myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2). Aberrant transcription and mRNA processing of multiple genes due to RNA-mediated toxic gain-of function has been suggested to cause the complex phenotype in DM1 and DM2. However, despite clinical and genetic similarities, DM1 and DM2 may be considered as distinct disorders. This review is an update on the latest findings specific to DM2, including explanations for the differences in clinical manifestations and pathophysiology between the two forms of myotonic dystrophies.
Collapse
Affiliation(s)
- Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, IRCCS Policlinico San Donato, Piazza E. Malan, 1, San Donato Mil., 20097, Milan, Italy. .,Department of Neurology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| |
Collapse
|
36
|
Larsen M, Kress W, Schoser B, Hehr U, Müller CR, Rost S. Identification of variants in MBNL1 in patients with a myotonic dystrophy-like phenotype. Eur J Hum Genet 2016; 24:1467-72. [PMID: 27222292 DOI: 10.1038/ejhg.2016.41] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 03/22/2016] [Accepted: 04/12/2016] [Indexed: 11/09/2022] Open
Abstract
The myotonic dystrophies (DMs) are the most common inherited muscular disorders in adults. In most of the cases, the disease is caused by (CTG)n/(CCTG)n repeat expansions (EXPs) in non-coding regions of the genes DMPK (dystrophia myotonica-protein kinase) and CNBP (CCHC-type zinc-finger nucleic acid-binding protein). The EXP is transcribed into mutant RNAs, which provoke a common pathomechanism that is characterized by misexpression and mis-splicing. In this study, we screened 138 patients with typical clinical features of DM being negative for EXP in the known genes. We sequenced DMPK and CNBP - associated with DM, as well as CELF1 (CUGBP, Elav-like family member 1) and MBNL1 (muscleblind-like splicing regulator 1) - associated with the pathomechanism of DM, for pathogenic variants, addressing the question whether defects in other genes could cause a DM-like phenotype. We identified variants in three unrelated patients in the MBNL1 gene, two of them were heterozygous missense mutations and one an in-frame deletion of three amino acids. The variants were located in different conserved regions of the protein. The missense mutations were classified as potentially pathogenic by prediction tools. Analysis of MBNL1 splice target genes was carried out for one of the patients using RNA from peripheral blood leukocytes (PBL). Analysis of six genes known to show mis-splicing in the skeletal muscle gave no informative results on the effect of this variant when tested in PBL. The association of these variants with the DM phenotype therefore remains unconfirmed, but we hope that in view of the key role of MBNL1 in DM pathogenesis our observations may foster further studies in this direction.
Collapse
Affiliation(s)
- Mirjam Larsen
- Department of Human Genetics, Julius-Maximilians-University, Würzburg, Germany
| | - Wolfram Kress
- Department of Human Genetics, Julius-Maximilians-University, Würzburg, Germany
| | - Benedikt Schoser
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Ute Hehr
- Center for and Department of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Clemens R Müller
- Department of Human Genetics, Julius-Maximilians-University, Würzburg, Germany
| | - Simone Rost
- Department of Human Genetics, Julius-Maximilians-University, Würzburg, Germany
| |
Collapse
|
37
|
Tan SV, Z'graggen WJ, Boërio D, Turner C, Hanna MG, Bostock H. In vivo assessment of muscle membrane properties in myotonic dystrophy. Muscle Nerve 2016; 54:249-57. [PMID: 26789642 DOI: 10.1002/mus.25025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/16/2015] [Accepted: 12/28/2015] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Myotonia in myotonic dystrophy types 1 (DM1) and 2 (DM2) is generally attributed to reduced chloride-channel conductance. We used muscle velocity recovery cycles (MVRCs) to investigate muscle membrane properties in DM1 and DM2, using comparisons with myotonia congenita (MC). METHODS MVRCs and responses to repetitive stimulation were compared between patients with DM1 (n = 18), DM2 (n = 5), MC (n = 18), and normal controls (n = 20). RESULTS Both DM1 and DM2 showed enhanced late supernormality after multiple conditioning stimuli, indicating delayed repolarization as in MC. Contrary to MC, however, DM1 showed reduced early supernormality after multiple conditioning stimuli, and weak DM1 patients also showed abnormally slow latency recovery after repetitive stimulation. CONCLUSIONS These findings support the presence of impaired chloride conductance in both DM1 and DM2. The early supernormality changes indicate that sodium currents were reduced in DM1, whereas the weakness-associated slow recovery after repetitive stimulation may provide an indication of reduced Na(+) /K(+) -ATPase activation. Muscle Nerve 54: 249-257, 2016.
Collapse
Affiliation(s)
- S Veronica Tan
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, The National, Queen Square, London, WC1N 3BG, UK.,Institute of Neurology, University College London, Queen Square, London, UK.,Department of Neurology and Neurophysiology, St Thomas' Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK.,Department of Academic Neurosciences, Kings College, London, UK
| | - Werner J Z'graggen
- Department of Neurosurgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Delphine Boërio
- Department of Neurosurgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Christopher Turner
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, The National, Queen Square, London, WC1N 3BG, UK.,Institute of Neurology, University College London, Queen Square, London, UK
| | - Michael G Hanna
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, The National, Queen Square, London, WC1N 3BG, UK.,Institute of Neurology, University College London, Queen Square, London, UK
| | - Hugh Bostock
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, The National, Queen Square, London, WC1N 3BG, UK.,Institute of Neurology, University College London, Queen Square, London, UK.,Department of Neurosurgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.,Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
38
|
Roles for RNA-binding proteins in development and disease. Brain Res 2016; 1647:1-8. [PMID: 26972534 DOI: 10.1016/j.brainres.2016.02.050] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022]
Abstract
RNA-binding protein activities are highly regulated through protein levels, intracellular localization, and post-translation modifications. During development, mRNA processing of specific gene sets is regulated through manipulation of functional RNA-binding protein activities. The impact of altered RNA-binding protein activities also affects human diseases in which there are either a gain-of-function or loss-of-function causes pathogenesis. We will discuss RNA-binding proteins and their normal developmental RNA metabolism and contrast how their function is disrupted in disease. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
|
39
|
Valaperta R, Lombardi F, Cardani R, Fossati B, Brigonzi E, Merli I, Sansone V, Merletti G, Spina E, Meola G, Costa E. Development and Validation of a New Molecular Diagnostic Assay for Detection of Myotonic Dystrophy Type 2. Genet Test Mol Biomarkers 2015; 19:703-9. [DOI: 10.1089/gtmb.2015.0135] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rea Valaperta
- Research Laboratories—Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
- Service of Laboratory Medicine, IRCCS Policlinico San Donato, Milan, Italy
| | - Fortunata Lombardi
- Research Laboratories—Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
- Service of Laboratory Medicine, IRCCS Policlinico San Donato, Milan, Italy
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
| | - Barbara Fossati
- Department of Biomedical Sciences for Health, University of Milan, IRCCS-Policlinico San Donato, Milan, Italy
| | - Elisa Brigonzi
- Department of Biomedical Sciences for Health, University of Milan, IRCCS-Policlinico San Donato, Milan, Italy
| | - Ilaria Merli
- Department of Biomedical Sciences for Health, University of Milan, IRCCS-Policlinico San Donato, Milan, Italy
| | - Valeria Sansone
- Neurorehabilitation Unit, University of Milan, NEMO Clinical Center (NeuroMuscular Omnicomprehensive), Fondazione Serena, Milan, Italy
| | - Giulia Merletti
- Research Laboratories—Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
| | - Edoardo Spina
- Research Laboratories—Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
| | - Giovanni Meola
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, IRCCS-Policlinico San Donato, Milan, Italy
| | - Elena Costa
- Research Laboratories—Molecular Biology, IRCCS Policlinico San Donato, Milan, Italy
- Service of Laboratory Medicine, IRCCS Policlinico San Donato, Milan, Italy
| |
Collapse
|
40
|
A Systematic Review and Meta-analysis on the Epidemiology of the Muscular Dystrophies. Can J Neurol Sci 2015; 43:163-77. [DOI: 10.1017/cjn.2015.311] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AbstractBackground: The muscular dystrophies are a heterogeneous group of genetic muscle diseases with variable distribution of weakness and mode of inheritance.Methods: We previously performed a systematic review of worldwide population-based studies on Duchenne and Becker muscular dystrophies; the current study focused on the epidemiology of other muscular dystrophies using Medline and EMBASE databases. Two reviewers independently reviewed all abstracts, full-text articles, and abstracted data from 1985 to 2011. Pooling of prevalence estimates was performed using random-effect models.Results: A total of 1104 abstracts and 167 full-text articles were reviewed. Thirty-one studies met all eligibility criteria and were included in the final analysis. The overall pooled prevalence of combined muscular dystrophies was 16.14 (confidence interval [CI], 11.21-23.23) per 100,000. The prevalence estimates per 100,000 were 8.26 (CI, 4.99-13.68) for myotonic dystrophy, 3.95 (CI, 2.89-5.40) for facioscapulohumeral dystrophy, 1.63 (CI, 0.94-2.81) for limb girdle muscular dystrophy, and 0.99 (CI, 0.62-1.57) for congenital muscular dystrophies.Conclusions: The studies differed widely in their approaches to case ascertainment, and substantial gaps remain in the global estimates of many other types of muscular dystrophies. Additional epidemiological studies using standardized diagnostic criteria as well as multiple sources of case ascertainment will help address the economic impact and health care burden of muscular dystrophies worldwide.
Collapse
|
41
|
Johnson NE, Hung M, Nasser E, Hagerman KA, Chen W, Ciafaloni E, Heatwole CR. The Impact of Pregnancy on Myotonic Dystrophy: A Registry-Based Study. J Neuromuscul Dis 2015; 2:447-452. [PMID: 27858748 PMCID: PMC5240614 DOI: 10.3233/jnd-150095] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The rate of symptom progression during pregnancy in myotonic dystrophy (DM) is not currently known. Further, there is little data regarding the rate of pregnancy complications and neonatal outcomes in DM. OBJECTIVE This study assesses symptom progression and complication rates during pregnancy in women with DM. METHODS DM women completed surveys regarding their prior pregnancies. Participants identified complications during their pregnancies and completed the Myotonic Dystrophy Health Index-Short Form (MDHI-SF) to measure their disease burden and identify the severity of select symptoms six-months prior to, during, and six-months after their first pregnancy. RESULTS 152 women with DM reported on 375 pregnancies. Among these pregnancies, there was a 32.5% miscarriage rate. Some complications were common including: pre-term labor (27.8%), pre-eclampsia (10.4%), and peripartum hemorrhage (13.9%). Participants' perception of their mobility and ability to perform activities, as measured by the MDHI-SF, worsened during pregnancy and did not recover during the post-partum period. DISCUSSION Miscarriage, maternal disease progression during pregnancy, and other pregnancy related complications may occur in DM. Women with DM should be counseled on these potential risks prior to considering pregnancy.
Collapse
Affiliation(s)
| | - Man Hung
- Department of Orthopaedics, The University of Utah, UT, USA
- Division of Epidemiology, The University of Utah, UT, USA
- Division of Public Health, The University of Utah, UT, USA
| | | | | | - Wei Chen
- Division of Epidemiology, The University of Utah, UT, USA
| | - Emma Ciafaloni
- The Department of Neurology, University of Rochester, NY, USA
| | | |
Collapse
|
42
|
Meola G, Cardani R. Myotonic Dystrophy Type 2: An Update on Clinical Aspects, Genetic and Pathomolecular Mechanism. J Neuromuscul Dis 2015; 2:S59-S71. [PMID: 27858759 PMCID: PMC5240594 DOI: 10.3233/jnd-150088] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Myotonic dystrophy (DM) is the most common adult muscular dystrophy, characterized by autosomal dominant progressive myopathy, myotonia and multiorgan involvement. To date two distinct forms caused by similar mutations have been identified. Myotonic dystrophy type 1 (DM1, Steinert's disease) is caused by a (CTG)n expansion in DMPK, while myotonic dystrophy type 2 (DM2) is caused by a (CCTG)n expansion in CNBP. Despite clinical and genetic similarities, DM1 and DM2 are distinct disorders. The pathogenesis of DM is explained by a common RNA gain-of-function mechanism in which the CUG and CCUG repeats alter cellular function, including alternative splicing of various genes. However additional pathogenic mechanism like changes in gene expression, modifier genes, protein translation and micro-RNA metabolism may also contribute to disease pathology and to clarify the phenotypic differences between these two types of myotonic dystrophies.This review is an update on the latest findings specific to DM2, including explanations for the differences in clinical manifestations and pathophysiology between the two forms of myotonic dystrophies.
Collapse
Affiliation(s)
- Giovanni Meola
- Department of Biomedical Sciences for Health, IRCCS Policlinico San Donato, University of Milan, San Donato Milanese, Milan, Italy
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| |
Collapse
|
43
|
Malatesta M, Cardani R, Pellicciari C, Meola G. RNA Transcription and Maturation in Skeletal Muscle Cells are Similarly Impaired in Myotonic Dystrophy and Sarcopenia: The Ultrastructural Evidence. Front Aging Neurosci 2014; 6:196. [PMID: 25126079 PMCID: PMC4115624 DOI: 10.3389/fnagi.2014.00196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 07/15/2014] [Indexed: 01/23/2023] Open
Affiliation(s)
- Manuela Malatesta
- Anatomy and Histology Section, Department of Neurological and Movement Sciences, University of Verona , Verona , Italy
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato , Milan , Italy
| | - Carlo Pellicciari
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology, University of Pavia , Pavia , Italy
| | - Giovanni Meola
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato , Milan , Italy ; Department of Neurology, University of Milan , Milan , Italy
| |
Collapse
|
44
|
Meola G, Cardani R. Myotonic dystrophies: An update on clinical aspects, genetic, pathology, and molecular pathomechanisms. Biochim Biophys Acta Mol Basis Dis 2014; 1852:594-606. [PMID: 24882752 DOI: 10.1016/j.bbadis.2014.05.019] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 01/18/2023]
Abstract
Myotonic dystrophy (DM) is the most common adult muscular dystrophy, characterized by autosomal dominant progressive myopathy, myotonia and multiorgan involvement. To date two distinct forms caused by similar mutations have been identified. Myotonic dystrophy type 1 (DM1, Steinert's disease) is caused by a (CTG)n expansion in DMPK, while myotonic dystrophy type 2 (DM2) is caused by a (CCTG)n expansion in ZNF9/CNBP. When transcribed into CUG/CCUG-containing RNA, mutant transcripts aggregate as nuclear foci that sequester RNA-binding proteins, resulting in spliceopathy of downstream effector genes. However, it is now clear that additional pathogenic mechanism like changes in gene expression, protein translation and micro-RNA metabolism may also contribute to disease pathology. Despite clinical and genetic similarities, DM1 and DM2 are distinct disorders requiring different diagnostic and management strategies. This review is an update on the recent advances in the understanding of the molecular mechanisms behind myotonic dystrophies. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.
Collapse
Affiliation(s)
- Giovanni Meola
- Department of Neurology, IRCCS Policlinico San Donato, University of Milan, San Donato Milanese, Milan, Italy; Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.
| |
Collapse
|
45
|
Hilbert JE, Ashizawa T, Day JW, Luebbe EA, Martens WB, McDermott MP, Tawil R, Thornton CA, Moxley RT. Diagnostic odyssey of patients with myotonic dystrophy. J Neurol 2013; 260:2497-504. [PMID: 23807151 PMCID: PMC4162528 DOI: 10.1007/s00415-013-6993-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 01/01/2023]
Abstract
The onset and symptoms of the myotonic dystrophies are diverse, complicating their diagnoses and limiting a comprehensive approach to their clinical care. This report analyzes the diagnostic delay (time from onset of first symptom to diagnosis) in a large sample of myotonic dystrophy (DM) patients enrolled in the US National Registry [679 DM type 1 (DM1) and 135 DM type 2 (DM2) patients]. Age of onset averaged 34.0 ± 14.1 years in DM2 patients compared to 26.1 ± 13.2 years in DM1 (p < 0.0001). The most common initial symptom in DM2 patients was leg weakness (32.6 %) compared to grip myotonia in DM1 (38.3 %). Pain was reported as the first symptom in 11.1 % of DM2 and 3.0 % of DM1 patients (p < 0.0001). Reaching the correct diagnosis in DM2 took 14 years on average (double the time compared to DM1) and a significantly higher percentage of patients underwent extended workup including electromyography, muscle biopsies, and finally genetic testing. DM patients who were index cases experienced similar diagnostic delays to non-index cases of DM. Further evaluation of how to shorten these diagnostic delays and limit their impact on burdens of disease, family planning, and symptom management is needed.
Collapse
Affiliation(s)
- James E Hilbert
- Department of Neurology, University of Rochester Medical Center, 601 Elmwood Ave, Box 673, Rochester, NY, 14642, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Malatesta M, Giagnacovo M, Costanzo M, Cisterna B, Cardani R, Meola G. Muscleblind-like1 undergoes ectopic relocation in the nuclei of skeletal muscles in myotonic dystrophy and sarcopenia. Eur J Histochem 2013; 57:e15. [PMID: 23807294 PMCID: PMC3794341 DOI: 10.4081/ejh.2013.e15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 01/24/2023] Open
Abstract
Muscleblind-like 1 (MBNL1) is an alternative splicing factor involved in postnatal development of skeletal muscles and heart in humans and mice, and its deregulation is known to be pivotal in the onset and development of myotonic dystrophy (DM). In fact, in DM patients this protein is ectopically sequestered into intranuclear foci, thus compromising the regulation of the alternative splicing of several genes. However, despite the numerous biochemical and molecular studies, scarce attention has been paid to the intranuclear location of MBNL1 outside the foci, although previous data demonstrated that in DM patients various splicing and cleavage factors undergo an abnormal intranuclear distribution suggestive of impaired RNA processing. Interestingly, these nuclear alterations strongly remind those observed in sarcopenia i.e., the loss of muscle mass and function which physiologically occurs during ageing. On this basis, in the present investigation the ultrastructural localization of MBNL1 was analyzed in the myonuclei of skeletal muscles from healthy and DM patients as well as from adult and old (sarcopenic) mice, in the attempt to elucidate possible changes in its distribution and amount. Our data demonstrate that in both dystrophic and sarcopenic muscles MBNL1 undergoes intranuclear relocation, accumulating in its usual functional sites but also ectopically moving to domains which are usually devoid of this protein in healthy adults. This accumulation/delocalization could contribute to hamper the functionality of the whole splicing machinery, leading to a lower nuclear metabolic activity and, consequently, to a less efficient protein synthesis. Moreover, the similar nuclear alterations found in DM and sarcopenia may account for the similar muscle tissue features (myofibre atrophy, fibre size variability and centrally located nuclei), and, in general, for the aging-reminiscent phenotype observed in DM patients.
Collapse
Affiliation(s)
- M Malatesta
- Dipartimento di Scienze Neurologiche, Neuropsicologiche, Morfologiche e Motorie, Sezione di Anatomia e Istologia, Università di Verona, 37134 Verona, Italy.
| | | | | | | | | | | |
Collapse
|
47
|
Modic M, Ule J, Sibley CR. CLIPing the brain: studies of protein-RNA interactions important for neurodegenerative disorders. Mol Cell Neurosci 2013; 56:429-35. [PMID: 23583633 PMCID: PMC3793874 DOI: 10.1016/j.mcn.2013.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/31/2013] [Accepted: 04/03/2013] [Indexed: 02/07/2023] Open
Abstract
The fate of an mRNA is largely determined by its interactions with RNA binding proteins (RBPs). Post-transcriptional processing, RNA stability, localisation and translation are some of the events regulated by the plethora of RBPs present within cells. Mutations in various RBPs cause several diseases of the central nervous system, including frontotemporal lobar degeneration, amyotrophic lateral sclerosis and fragile X syndrome. Here we review the studies that integrated UV-induced cross-linked immunoprecipitation (CLIP) with other genome-wide methods to comprehensively characterise the function of diverse RBPs in the brain. We discuss the technical challenges of these studies and review the strategies that can be used to reliably identify the RNAs bound and regulated by an RBP. We conclude by highlighting how CLIP and related techniques have been instrumental in addressing the role of RBPs in neurologic diseases. This article is part of a Special Issue entitled: RNA and splicing regulation in neurodegeneration.
Collapse
Affiliation(s)
- Miha Modic
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge, CB2 0QH, UK
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377 Munich, Germany
| | - Jernej Ule
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge, CB2 0QH, UK
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Corresponding authors at: Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK. Fax: + 44 1223213556.
| | - Christopher R. Sibley
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge, CB2 0QH, UK
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Corresponding authors at: Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK. Fax: + 44 1223213556.
| |
Collapse
|
48
|
Heatwole CR, Statland JM, Logigian EL. The diagnosis and treatment of myotonic disorders. Muscle Nerve 2013; 47:632-48. [PMID: 23536309 DOI: 10.1002/mus.23683] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2012] [Indexed: 12/12/2022]
Abstract
Myotonia is a defining clinical symptom and sign common to a relatively small group of muscle diseases, including the myotonic dystrophies and the nondystrophic myotonic disorders. Myotonia can be observed on clinical examination, as can its electrical correlate, myotonic discharges, on electrodiagnostic testing. Research interest in the myotonic disorders continues to expand rapidly, which justifies a review of the scientific bases, clinical manifestations, and numerous therapeutic approaches associated with these disorders. We review the pathomechanisms of myotonia, the clinical features of the dystrophic and nondystrophic myotonic disorders, and the diagnostic approach and treatment options for patients with symptomatic myotonia.
Collapse
Affiliation(s)
- Chad R Heatwole
- Department of Neurology, Box 673, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, New York 14642, USA.
| | | | | |
Collapse
|
49
|
Singh M. Dysregulated A to I RNA editing and non-coding RNAs in neurodegeneration. Front Genet 2013; 3:326. [PMID: 23346095 PMCID: PMC3551214 DOI: 10.3389/fgene.2012.00326] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 12/28/2012] [Indexed: 12/14/2022] Open
Abstract
RNA editing is an alteration in the primary nucleotide sequences resulting from a chemical change in the base. RNA editing is observed in eukaryotic mRNA, transfer RNA, ribosomal RNA, and non-coding RNAs (ncRNA). The most common RNA editing in the mammalian central nervous system is a base modification, where the adenosine residue is base-modified to inosine (A to I). Studies from ADAR (adenosine deaminase that act on RNA) mutants in Caenorhabditis elegans, Drosophila, and mice clearly show that the RNA editing process is an absolute requirement for nervous system homeostasis and normal physiology of the animal. Understanding the mechanisms of editing and findings of edited substrates has provided a better knowledge of the phenotype due to defective and hyperactive RNA editing. A to I RNA editing is catalyzed by a family of enzymes knows as ADARs. ADARs modify duplex RNAs and editing of duplex RNAs formed by ncRNAs can impact RNA functions, leading to an altered regulatory gene network. Such altered functions by A to I editing is observed in mRNAs, microRNAs (miRNA) but other editing of small and long ncRNAs (lncRNAs) has yet to be identified. Thus, ncRNA and RNA editing may provide key links between neural development, nervous system function, and neurological diseases. This review includes a summary of seminal findings regarding the impact of ncRNAs on biological and pathological processes, which may be further modified by RNA editing. NcRNAs are non-translated RNAs classified by size and function. Known ncRNAs like miRNAs, smallRNAs (smRNAs), PIWI-interacting RNAs (piRNAs), and lncRNAs play important roles in splicing, DNA methylation, imprinting, and RNA interference. Of note, miRNAs are involved in development and function of the nervous system that is heavily dependent on both RNA editing and the intricate spatiotemporal expression of ncRNAs. This review focuses on the impact of dysregulated A to I editing and ncRNAs in neurodegeneration.
Collapse
Affiliation(s)
- Minati Singh
- Department of Internal Medicine, University of Iowa Iowa City, IA, USA
| |
Collapse
|
50
|
Fratta P, Mizielinska S, Nicoll AJ, Zloh M, Fisher EMC, Parkinson G, Isaacs AM. C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci Rep 2012; 2:1016. [PMID: 23264878 PMCID: PMC3527825 DOI: 10.1038/srep01016] [Citation(s) in RCA: 250] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/07/2012] [Indexed: 12/14/2022] Open
Abstract
Large expansions of a non-coding GGGGCC-repeat in the first intron of the C9orf72 gene are a common cause of both amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). G-rich sequences have a propensity for forming highly stable quadruplex structures in both RNA and DNA termed G-quadruplexes. G-quadruplexes have been shown to be involved in a range of processes including telomere stability and RNA transcription, splicing, translation and transport. Here we show using NMR and CD spectroscopy that the C9orf72 hexanucleotide expansion can form a stable G-quadruplex, which has profound implications for disease mechanism in ALS and FTD.
Collapse
Affiliation(s)
- Pietro Fratta
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | | | | | | | | | |
Collapse
|