1
|
Sane H, Nivins S, Paranjape A, Gokulchandran N, Badhe S, Varghese R, Badhe P, Sharma A. Severity of muscle impairment and its progression assessed using musculoskeletal magnetic resonance imaging and diffusion tension imaging in 78 boys with Duchenne muscular dystrophy: a retrospective study. Pol J Radiol 2024; 89:e88-e105. [PMID: 38510548 PMCID: PMC10953512 DOI: 10.5114/pjr.2024.135718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/19/2024] [Indexed: 03/22/2024] Open
Abstract
Purpose Duchenne muscular dystrophy (DMD) is the most common and severe form of muscular dystrophy. Current diagnostic tests like genetic testing, needle electromyography, and muscle biopsy are either not easily available or invasive, and they are impractical for assessing disease progression and treatment outcomes. Therefore, there is a need for a non-invasive and accurate investigative modality for DMD. In recent years, musculoskeletal magnetic resonance imaging (MRI-MSK) along with fractional anisotropy (FA) and diffusion tensor imaging (DTI) have become major non-invasive tools. Material and methods T1-weighted MRI-MSK and FA measures of DTI of 78 DMD patients were retrospectively studied to identify the distinct pattern of muscle involvement and fatty infiltration as age and/or disease progresses. Correlation analysis was performed between MRI-MSK grade score vs. age, muscle strength, and Vignos scale. Spearman's rank correlation coefficient was used. Results As age increased, the MRI grade score and Vignos score increased. There was a statistically significant high positive correlation between MRI-MSK grade score and age, and low positive correlation with Vignos scores. With increasing age, the muscle strength on manual muscle testing (MMT) and FA value decreased. There was high negative correlation with muscle strength on MMT and low positive correlation between FA values and MMT score. Conclusions On T1-weighted MRI, a distinct pattern, extent, and distribution of lower limb muscle involvement can be seen. MRI-MSK grade score worsens with progressing age, reducing strength, and increasing functional impairment. FA alone may not be an accurate marker in assessing progression of DMD. MRI-MSK and other DTI measures should be further explored as diagnostic and prognostic tools for DMD.
Collapse
Affiliation(s)
- Hemangi Sane
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Samson Nivins
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Amruta Paranjape
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Nandini Gokulchandran
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Suvarna Badhe
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Ritu Varghese
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Prerna Badhe
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| | - Alok Sharma
- NeuroGen Brain and Spine Institute, Stemasia Hospital and Research Centre, Navi Mumbai , Maharashtra, India
| |
Collapse
|
2
|
Hiyoshi T, Zhao F, Baba R, Hirakawa T, Kuboki R, Suzuki K, Tomimatsu Y, O'Donnell P, Han S, Zach N, Nakashima M. Electrical impedance myography detects dystrophin-related muscle changes in mdx mice. Skelet Muscle 2023; 13:19. [PMID: 37980539 PMCID: PMC10657153 DOI: 10.1186/s13395-023-00331-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND The lack of functional dystrophin protein in Duchenne muscular dystrophy (DMD) causes chronic skeletal muscle inflammation and degeneration. Therefore, the restoration of functional dystrophin levels is a fundamental approach for DMD therapy. Electrical impedance myography (EIM) is an emerging tool that provides noninvasive monitoring of muscle conditions and has been suggested as a treatment response biomarker in diverse indications. Although magnetic resonance imaging (MRI) of skeletal muscles has become a standard measurement in clinical trials for DMD, EIM offers distinct advantages, such as portability, user-friendliness, and reduced cost, allowing for remote monitoring of disease progression or response to therapy. To investigate the potential of EIM as a biomarker for DMD, we compared longitudinal EIM data with MRI/histopathological data from an X-linked muscular dystrophy (mdx) mouse model of DMD. In addition, we investigated whether EIM could detect dystrophin-related changes in muscles using antisense-mediated exon skipping in mdx mice. METHODS The MRI data for muscle T2, the magnetic resonance spectroscopy (MRS) data for fat fraction, and three EIM parameters with histopathology were longitudinally obtained from the hindlimb muscles of wild-type (WT) and mdx mice. In the EIM study, a cell-penetrating peptide (Pip9b2) conjugated antisense phosphorodiamidate morpholino oligomer (PPMO), designed to induce exon-skipping and restore functional dystrophin production, was administered intravenously to mdx mice. RESULTS MRI imaging in mdx mice showed higher T2 intensity at 6 weeks of age in hindlimb muscles compared to WT mice, which decreased at ≥ 9 weeks of age. In contrast, EIM reactance began to decline at 12 weeks of age, with peak reduction at 18 weeks of age in mdx mice. This decline was associated with myofiber atrophy and connective tissue infiltration in the skeletal muscles. Repeated dosing of PPMO (10 mg/kg, 4 times every 2 weeks) in mdx mice led to an increase in muscular dystrophin protein and reversed the decrease in EIM reactance. CONCLUSIONS These findings suggest that muscle T2 MRI is sensitive to the early inflammatory response associated with dystrophin deficiency, whereas EIM provides a valuable biomarker for the noninvasive monitoring of subsequent changes in skeletal muscle composition. Furthermore, EIM reactance has the potential to monitor dystrophin-deficient muscle abnormalities and their recovery in response to antisense-mediated exon skipping.
Collapse
Affiliation(s)
- Tetsuaki Hiyoshi
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Fuqiang Zhao
- Center of Excellence for Imaging, Preclinical and Translational Sciences, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Rina Baba
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Takeshi Hirakawa
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Ryosuke Kuboki
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Kazunori Suzuki
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yoshiro Tomimatsu
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Patricio O'Donnell
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Steve Han
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Neta Zach
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Masato Nakashima
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| |
Collapse
|
3
|
Martín-Noguerol T, Barousse R, Wessell DE, Rossi I, Luna A. Clinical applications of skeletal muscle diffusion tensor imaging. Skeletal Radiol 2023; 52:1639-1649. [PMID: 37083977 DOI: 10.1007/s00256-023-04350-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Diffusion tensor imaging (DTI) may allow the determination of new threshold values, based on water anisotropy, to differentiate between healthy muscle and various pathological processes. Additionally, it may quantify treatment monitoring or training effects. Most current studies have evaluated the potential of DTI of skeletal muscle to assess sports-related injuries or therapy, and training monitoring. Another critical area of application of this technique is the characterization and monitoring of primary and secondary myopathies. In this manuscript, we review the application of DTI in the evaluation of skeletal muscle in these and other novel clinical scenarios, with emphasis on the use of quantitative imaging-derived biomarkers. Finally, the main limitations of the introduction of DTI in the clinical setting and potential areas of future use are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Luna
- MRI Unit, Radiology Department, HT Médica, Jaén, Spain
| |
Collapse
|
4
|
Pedersen JJ, Stemmerik MG, Jacobsen LN, Skriver SV, Wilms GR, Duno M, Vissing J. Muscle fat replacement and contractility in patients with skeletal muscle sodium channel disorders. Sci Rep 2023; 13:2538. [PMID: 36782059 PMCID: PMC9925746 DOI: 10.1038/s41598-023-29759-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Skeletal muscle sodium channel disorders give rise to episodic symptoms such as myotonia and/or periodic paralysis. Chronic symptoms with permanent weakness are not considered characteristic of the phenotypes. Muscle fat replacement represents irreversible damage that inevitably will impact on muscle strength. This study investigates muscle fat replacement and contractility in patients with pathogenic SCN4A variants compared to healthy controls. T1-weighted and 2-point Dixon MRI of the legs were conducted to assess fat replacement. Stationary dynamometry was used to assess muscle strength. Contractility was determined by maximal muscle contraction divided by cross-sectional muscle area. The average cross-sectional intramuscular fat fraction was greater in patients compared with controls by 2.5% in the calves (95% CI 0.74-4.29%, p = 0.007) and by 2.0% in the thighs (95% CI 0.75-3.2%, p = 0.003). Muscle contractility was less in patients vs. controls by 14-27% (p < 0.05). Despite greater fat fraction and less contractility, absolute strength was not significantly less. This study quantitatively documents greater fat fraction and additionally describes difference in muscle contractility in a large cohort of patients with skeletal muscle sodium channel disorders. The clinical impact of these abnormal findings is likely limited as muscle hypertrophy in the patients served to preserve absolute muscle strength. Subgroup analysis indicated significant difference in phenotype by genotype, however these findings lack statistical significance and serve as inspiration for future researchers to probe into the geno- phenotype relationship in these disorders.Trial registration: The study was registered at http://clinicaltrials.gov (identifier: NCT04808388).
Collapse
Affiliation(s)
- Jonas Jalili Pedersen
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Inge Lehmanns Vej 7-9, 2100, Copenhagen, Denmark.
| | - Mads Godtfeldt Stemmerik
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Inge Lehmanns Vej 7-9, 2100, Copenhagen, Denmark
| | - Laura Nørager Jacobsen
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Inge Lehmanns Vej 7-9, 2100, Copenhagen, Denmark
| | - Sofie Vinther Skriver
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Inge Lehmanns Vej 7-9, 2100, Copenhagen, Denmark
| | - Gustav Rhode Wilms
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Inge Lehmanns Vej 7-9, 2100, Copenhagen, Denmark
| | - Morten Duno
- Clinical Genetic Laboratory, Department of Biochemical Genetics, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Inge Lehmanns Vej 7-9, 2100, Copenhagen, Denmark
| |
Collapse
|
5
|
Cristiano L, Brogna C, Tasca G, Verdolotti T, Pane M, Mercuri E. Muscle-MRI and Functional Levels for the Evaluation of Upper Limbs in Duchenne Muscular Dystrophy: A Critical Review of the Literature. Medicina (B Aires) 2022; 58:medicina58030440. [PMID: 35334617 PMCID: PMC8954550 DOI: 10.3390/medicina58030440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/02/2022] Open
Abstract
Many qualitative and quantitative Magnetic Resonance Imaging (MRI) techniques have been applied to evaluate muscle fat degeneration in Duchenne muscular dystrophy (DMD) subjects, but only few studies have focused on the upper limbs. We reviewed the literature in order to evaluate the association between muscle MRI findings and motor function levels in the upper limbs of DMD patients. Ten studies with upper limb muscle MRI data were available. Four explored all upper limb segments, while six explored only the forearm. Functional assessments were performed in nine of the ten studies. All of the studies showed a significant correlation between muscle MRI changes and motor function levels in both ambulant and non-ambulant DMD patients.
Collapse
Affiliation(s)
- Lara Cristiano
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
| | - Claudia Brogna
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-30155340; Fax: +39-06-30154363
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
| | - Tommaso Verdolotti
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
| | - Marika Pane
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
6
|
Oh HJ, Chung E, Kim J, Kim MJ, Kim GA, Lee SH, Ra K, Eom K, Park S, Chae JH, Kim JS, Lee BC. Generation of a Dystrophin Mutant in Dog by Nuclear Transfer Using CRISPR/Cas9-Mediated Somatic Cells: A Preliminary Study. Int J Mol Sci 2022; 23:ijms23052898. [PMID: 35270040 PMCID: PMC8911381 DOI: 10.3390/ijms23052898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Dystrophinopathy is caused by mutations in the dystrophin gene, which lead to progressive muscle degeneration, necrosis, and finally, death. Recently, golden retrievers have been suggested as a useful animal model for studying human dystrophinopathy, but the model has limitations due to difficulty in maintaining the genetic background using conventional breeding. In this study, we successfully generated a dystrophin mutant dog using the CRISPR/Cas9 system and somatic cell nuclear transfer. The dystrophin mutant dog displayed phenotypes such as elevated serum creatine kinase, dystrophin deficiency, skeletal muscle defects, an abnormal electrocardiogram, and avoidance of ambulation. These results indicate that donor cells with CRISPR/Cas9 for a specific gene combined with the somatic cell nuclear transfer technique can efficiently produce a dystrophin mutant dog, which will help in the successful development of gene therapy drugs for dogs and humans.
Collapse
Affiliation(s)
- Hyun Ju Oh
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.J.O.); (M.J.K.); (G.A.K.); (S.H.L.); (K.R.)
| | - Eugene Chung
- Center for Genome Engineering, Institute for Basic Science, Seoul 08826, Korea;
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Jaehwan Kim
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul 5029, Korea; (J.K.); (K.E.)
| | - Min Jung Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.J.O.); (M.J.K.); (G.A.K.); (S.H.L.); (K.R.)
| | - Geon A. Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.J.O.); (M.J.K.); (G.A.K.); (S.H.L.); (K.R.)
- Department of Clinical Pathology, College of Health Science, Eulji University, Uijeongbu 11759, Korea
| | - Seok Hee Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.J.O.); (M.J.K.); (G.A.K.); (S.H.L.); (K.R.)
| | - Kihae Ra
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.J.O.); (M.J.K.); (G.A.K.); (S.H.L.); (K.R.)
| | - Kidong Eom
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul 5029, Korea; (J.K.); (K.E.)
| | - Soojin Park
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (S.P.); (J.-H.C.)
| | - Jong-Hee Chae
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (S.P.); (J.-H.C.)
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul 08826, Korea;
- Correspondence: (J.-S.K.); (B.C.L.); Tel.: +82-2-880-9327 (J.-S.K.); +82-2-880-1269 (B.C.L.)
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.J.O.); (M.J.K.); (G.A.K.); (S.H.L.); (K.R.)
- Correspondence: (J.-S.K.); (B.C.L.); Tel.: +82-2-880-9327 (J.-S.K.); +82-2-880-1269 (B.C.L.)
| |
Collapse
|
7
|
Bell M, Al Masruri G, Fernandez J, Williams SA, Agur AM, Stott NS, Hajarizadeh B, Mirjalili A. Typical m. triceps surae morphology and architecture measurement from 0 to 18 years: A narrative review. J Anat 2021; 240:746-760. [PMID: 34750816 PMCID: PMC8930835 DOI: 10.1111/joa.13584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 12/01/2022] Open
Abstract
The aim of this review was to report on the imaging modalities used to assess morphological and architectural properties of the m. triceps surae muscle in typically developing children, and the available reliability analyses. Scopus and MEDLINE (Pubmed) were searched systematically for all original articles published up to September 2020 measuring morphological and architectural properties of the m. triceps surae in typically developing children (18 years or under). Thirty eligible studies were included in this analysis, measuring fibre bundle length (FBL) (n = 11), pennation angle (PA) (n = 10), muscle volume (MV) (n = 16) and physiological cross‐sectional area (PCSA) (n = 4). Three primary imaging modalities were utilised to assess these architectural parameters in vivo: two‐dimensional ultrasound (2DUS; n = 12), three‐dimensional ultrasound (3DUS; n = 9) and magnetic resonance imaging (MRI; n = 6). The mean age of participants ranged from 1.4 years to 18 years old. There was an apparent increase in m. gastrocnemius medialis MV and pCSA with age; however, no trend was evident with FBL or PA. Analysis of correlations of muscle variables with age was limited by a lack of longitudinal data and methodological variations between studies affecting outcomes. Only five studies evaluated the reliability of the methods. Imaging methodologies such as MRI and US may provide valuable insight into the development of skeletal muscle from childhood to adulthood; however, variations in methodological approaches can significantly influence outcomes. Researchers wishing to develop a model of typical muscle development should carry out longitudinal architectural assessment of all muscles comprising the m. triceps surae utilising a consistent approach that minimises confounding errors.
Collapse
Affiliation(s)
- Matthew Bell
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ghaliya Al Masruri
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Justin Fernandez
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | - Sîan A Williams
- Faculty of Health Sciences, Curtin School of Allied Health, Curtin University, Perth, Australia.,Faculty of Medical and Health Sciences, Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Anne M Agur
- Division of Anatomy, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Ngaire S Stott
- Faculty of Medical and Health Sciences, Department of Surgery, University of Auckland, Auckland, New Zealand
| | | | - Ali Mirjalili
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
8
|
Musculoskeletal magnetic resonance imaging in the DE50-MD dog model of Duchenne muscular dystrophy. Neuromuscul Disord 2021; 31:736-751. [PMID: 34384671 PMCID: PMC8449064 DOI: 10.1016/j.nmd.2021.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022]
Abstract
Normalized muscle volumes distinguish between wild type and DE50-MD dogs. Global muscle T2 signal intensities discriminate between wild type and DE50-MD dogs. Musculoskeletal changes detected by MRI reflect those seen in human DMD patients. Musculoskeletal MRI in this model will be useful to assess treatment efficacy.
The DE50-MD canine model of Duchenne muscular dystrophy (DMD) has a dystrophin gene splice site mutation causing deletion of exon 50, an out-of-frame transcript and absence of dystrophin expression in striated muscles. We hypothesized that the musculoskeletal phenotype of DE50-MD dogs could be detected using Magnetic Resonance Imaging (MRI), that it would progress with age and that it would reflect those in other canine models and DMD patients. 15 DE50-MD and 10 age-matched littermate wild type (WT) male dogs underwent MRI every 3 months from 3 to 18 months of age. Normalized muscle volumes, global muscle T2 and ratio of post- to pre-gadolinium T1-weighted SI were evaluated in 7 pelvic limb and 4 lumbar muscles bilaterally. DE50-MD dogs, compared to WT, had smaller volumes in all muscles, except the cranial sartorius; global muscle T2 was significantly higher in DE50-MD dogs compared to WT. Muscle volumes plateaued and global muscle T2 decreased with age. Normalized muscle volumes and global muscle T2 revealed significant differences between groups longitudinally and should be useful to determine efficacy of therapeutics in this model with suitable power and low sample sizes. Musculoskeletal changes reflect those of DMD patients and other dog models.
Collapse
|
9
|
Lee CM, Kang BK, Kim M. Radiologic Definition of Sarcopenia in Chronic Liver Disease. Life (Basel) 2021; 11:86. [PMID: 33504046 PMCID: PMC7910987 DOI: 10.3390/life11020086] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Sarcopenia is prevalent in patients with chronic liver disease, and affected patients tend to have worse clinical outcomes and higher mortality. However, relevant analyses are limited by heterogeneity in the definition of sarcopenia and in the methodological approaches in assessing it. We reviewed several radiologic methods for sarcopenia in patients with chronic liver disease. Dual energy X-ray absorptiometry (DXA) can measure muscle mass, but it is difficult to evaluate muscle quality using this technique. Computed tomography, known as the gold standard for diagnosing sarcopenia, enables the objective measurement of muscle quantity and quality. The third lumbar skeletal muscle index (L3 SMI) more accurately predicted the mortality of subjects than the psoas muscle index (PMI). Few studies have evaluated the sarcopenia of chronic liver disease using ultrasonography and magnetic resonance imaging, and more studies are needed. Unification of the measurement method and cut-off value would facilitate a more systematic and universal prognosis evaluation in patients with chronic liver disease.
Collapse
Affiliation(s)
| | | | - Mimi Kim
- Department of Radiology, College of Medicine, Hanyang University, Seoul 04763, Korea; (C.-m.L.); (B.K.K.)
| |
Collapse
|
10
|
Radiologic Definition of Sarcopenia in Chronic Liver Disease. LIFE (BASEL, SWITZERLAND) 2021. [PMID: 33504046 DOI: 10.3390/life11020086.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Sarcopenia is prevalent in patients with chronic liver disease, and affected patients tend to have worse clinical outcomes and higher mortality. However, relevant analyses are limited by heterogeneity in the definition of sarcopenia and in the methodological approaches in assessing it. We reviewed several radiologic methods for sarcopenia in patients with chronic liver disease. Dual energy X-ray absorptiometry (DXA) can measure muscle mass, but it is difficult to evaluate muscle quality using this technique. Computed tomography, known as the gold standard for diagnosing sarcopenia, enables the objective measurement of muscle quantity and quality. The third lumbar skeletal muscle index (L3 SMI) more accurately predicted the mortality of subjects than the psoas muscle index (PMI). Few studies have evaluated the sarcopenia of chronic liver disease using ultrasonography and magnetic resonance imaging, and more studies are needed. Unification of the measurement method and cut-off value would facilitate a more systematic and universal prognosis evaluation in patients with chronic liver disease.
Collapse
|
11
|
Pandeya SR, Nagy JA, Riveros D, Semple C, Taylor RS, Mortreux M, Sanchez B, Kapur K, Rutkove SB. Predicting myofiber cross-sectional area and triglyceride content with electrical impedance myography: A study in db/db mice. Muscle Nerve 2021; 63:127-140. [PMID: 33063867 PMCID: PMC8891989 DOI: 10.1002/mus.27095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/02/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Electrical impedance myography (EIM) provides insight into muscle composition and structure. We sought to evaluate its use in a mouse obesity model characterized by myofiber atrophy. METHODS We applied a prediction algorithm, ie, the least absolute shrinkage and selection operator (LASSO), to surface, needle array, and ex vivo EIM data from db/db and wild-type mice and assessed myofiber cross-sectional area (CSA) histologically and triglyceride (TG) content biochemically. RESULTS EIM data from all three modalities provided acceptable predictions of myofiber CSA with average root mean square error (RMSE) of 15% in CSA (ie, ±209 μm2 for a mean CSA of 1439 μm2 ) and TG content with RMSE of 30% in TG content (ie, ±7.3 nmol TG/mg muscle for a mean TG content of 25.4 nmol TG/mg muscle). CONCLUSIONS EIM combined with a predictive algorithm provides reasonable estimates of myofiber CSA and TG content without the need for biopsy.
Collapse
Affiliation(s)
- Sarbesh R. Pandeya
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Janice A. Nagy
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Daniela Riveros
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Carson Semple
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Rebecca S. Taylor
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Marie Mortreux
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Benjamin Sanchez
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, Utah
| | - Kush Kapur
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Seward B. Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Gao S, Chen SN, Di Nardo C, Lombardi R. Arrhythmogenic Cardiomyopathy and Skeletal Muscle Dystrophies: Shared Histopathological Features and Pathogenic Mechanisms. Front Physiol 2020; 11:834. [PMID: 32848821 PMCID: PMC7406798 DOI: 10.3389/fphys.2020.00834] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable cardiac disease characterized by fibrotic or fibrofatty myocardial replacement, associated with an increased risk of ventricular arrhythmias and sudden cardiac death. Originally described as a disease of the right ventricle, ACM is currently recognized as a biventricular entity, due to the increasing numbers of reports of predominant left ventricular or biventricular involvement. Research over the last 20 years has significantly advanced our knowledge of the etiology and pathogenesis of ACM. Several etiopathogenetic theories have been proposed; among them, the most attractive one is the dystrophic theory, based on the observation of similar histopathological features between ACM and skeletal muscle dystrophies (SMDs), such as progressive muscular degeneration, inflammation, and tissue replacement by fatty and fibrous tissue. This review will describe the pathophysiological and molecular similarities shared by ACM with SMDs.
Collapse
Affiliation(s)
- Shanshan Gao
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Suet Nee Chen
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Carlo Di Nardo
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Raffaella Lombardi
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO, United States.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| |
Collapse
|
13
|
Zabaleta-Korta A, Fernández-Peña E, Santos-Concejero J. Regional Hypertrophy, the Inhomogeneous Muscle Growth: A Systematic Review. Strength Cond J 2020. [DOI: 10.1519/ssc.0000000000000574] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
14
|
Rooney WD, Berlow YA, Triplett WT, Forbes SC, Willcocks RJ, Wang DJ, Arpan I, Arora H, Senesac C, Lott DJ, Tennekoon G, Finkel R, Russman BS, Finanger EL, Chakraborty S, O'Brien E, Moloney B, Barnard A, Sweeney HL, Daniels MJ, Walter GA, Vandenborne K. Modeling disease trajectory in Duchenne muscular dystrophy. Neurology 2020; 94:e1622-e1633. [PMID: 32184340 DOI: 10.1212/wnl.0000000000009244] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/17/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To quantify disease progression in individuals with Duchenne muscular dystrophy (DMD) using magnetic resonance biomarkers of leg muscles. METHODS MRI and magnetic resonance spectroscopy (MRS) biomarkers were acquired from 104 participants with DMD and 51 healthy controls using a prospective observational study design with patients with DMD followed up yearly for up to 6 years. Fat fractions (FFs) in vastus lateralis and soleus muscles were determined with 1H MRS. MRI quantitative T2 (qT2) values were measured for 3 muscles of the upper leg and 5 muscles of the lower leg. Longitudinal changes in biomarkers were modeled with a cumulative distribution function using a nonlinear mixed-effects approach. RESULTS MRS FF and MRI qT2 increased with DMD disease duration, with the progression time constants differing markedly between individuals and across muscles. The average age at half-maximal muscle involvement (μ) occurred 4.8 years earlier in vastus lateralis than soleus, and these measures were strongly associated with loss-of-ambulation age. Corticosteroid treatment was found to delay μ by 2.5 years on average across muscles, although there were marked differences between muscles with more slowly progressing muscles showing larger delay. CONCLUSIONS MRS FF and MRI qT2 provide sensitive noninvasive measures of DMD progression. Modeling changes in these biomarkers across multiple muscles can be used to detect and monitor the therapeutic effects of corticosteroids on disease progression and to provide prognostic information on functional outcomes. This modeling approach provides a method to transform these MRI biomarkers into well-understood metrics, allowing concise summaries of DMD disease progression at individual and population levels. CLINICALTRIALSGOV IDENTIFIER NCT01484678.
Collapse
Affiliation(s)
- William D Rooney
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR.
| | - Yosef A Berlow
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - William T Triplett
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Sean C Forbes
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Rebecca J Willcocks
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Dah-Jyuu Wang
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Ishu Arpan
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Harneet Arora
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Claudia Senesac
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Donovan J Lott
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Gihan Tennekoon
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Richard Finkel
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Barry S Russman
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Erika L Finanger
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Saptarshi Chakraborty
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Elliott O'Brien
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Brendan Moloney
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Alison Barnard
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - H Lee Sweeney
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Michael J Daniels
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Glenn A Walter
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Krista Vandenborne
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| |
Collapse
|
15
|
Ropars J, Gravot F, Ben Salem D, Rousseau F, Brochard S, Pons C. Muscle MRI: A biomarker of disease severity in Duchenne muscular dystrophy? A systematic review. Neurology 2019; 94:117-133. [PMID: 31892637 DOI: 10.1212/wnl.0000000000008811] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To assess the evidence of a relationship between muscle MRI and disease severity in Duchenne muscular dystrophy (DMD). METHODS We conducted a systematic review of studies that analyzed correlations between MRI measurements and motor function in patients with DMD. PubMed, Cochrane, Scopus, and Web of Science were searched using relevant keywords and inclusion/exclusion criteria (January 1, 1990-January 31, 2019). We evaluated article quality using the Joanna Briggs Institute scale. Information regarding the samples included, muscles evaluated, MRI protocols and motor function tests used was collected from each article. Correlations between MRI measurements and motor function were reported exhaustively. RESULTS Seventeen of 1,629 studies identified were included. Most patients included were ambulant with a mean age of 8.9 years. Most studies evaluated lower limb muscles. Moderate to excellent correlations were found between MRI measurements and motor function. The strongest correlations were found for quantitative MRI measurements such as fat fraction or mean T2. Correlations were stronger for lower leg muscles such as soleus. One longitudinal study reported that changes in soleus mean T2 were highly correlated with changes in motor function. CONCLUSION The findings of this systematic review showed that MRI measurements can be used as biomarkers of disease severity in ambulant patients with DMD. Guidelines are proposed to help clinicians choose the most appropriate MRI measurements and muscles to evaluate. Studies exploring upper limb muscles, other stages of the disease, and sensitivity of measurements to change are needed.
Collapse
Affiliation(s)
- Juliette Ropars
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France.
| | - France Gravot
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Douraied Ben Salem
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - François Rousseau
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Sylvain Brochard
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Christelle Pons
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| |
Collapse
|
16
|
Krishnamurthy R, Wang DJJ, Cervantes B, McAllister A, Nelson E, Karampinos DC, Hu HH. Recent Advances in Pediatric Brain, Spine, and Neuromuscular Magnetic Resonance Imaging Techniques. Pediatr Neurol 2019; 96:7-23. [PMID: 31023603 DOI: 10.1016/j.pediatrneurol.2019.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 02/25/2019] [Accepted: 03/03/2019] [Indexed: 12/21/2022]
Abstract
Magnetic resonance imaging (MRI) is a powerful radiologic tool with the ability to generate a variety of proton-based signal contrast from tissues. Owing to this immense flexibility in signal generation, new MRI techniques are constantly being developed, tested, and optimized for clinical utility. In addition, the safe and nonionizing nature of MRI makes it a suitable modality for imaging in children. In this review article, we summarize a few of the most popular advances in MRI techniques in recent years. In particular, we highlight how these new developments have affected brain, spine, and neuromuscular imaging and focus on their applications in pediatric patients. In the first part of the review, we discuss new approaches such as multiphase and multidelay arterial spin labeling for quantitative perfusion and angiography of the brain, amide proton transfer MRI of the brain, MRI of brachial plexus and lumbar plexus nerves (i.e., neurography), and T2 mapping and fat characterization in neuromuscular diseases. In the second part of the review, we focus on describing new data acquisition strategies in accelerated MRI aimed collectively at reducing the scan time, including simultaneous multislice imaging, compressed sensing, synthetic MRI, and magnetic resonance fingerprinting. In discussing the aforementioned, the review also summarizes the advantages and disadvantages of each method and their current state of commercial availability from MRI vendors.
Collapse
Affiliation(s)
| | - Danny J J Wang
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Barbara Cervantes
- Department of Diagnostic and Interventional Radiology, Technische Universität München, Munich, Germany
| | | | - Eric Nelson
- Center for Biobehavioral Health, Nationwide Children's Hospital, Columbus, Ohio
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, Technische Universität München, Munich, Germany
| | | |
Collapse
|
17
|
Effectiveness of High-Speed T2-Corrected Multiecho MR Spectroscopic Method for Quantifying Thigh Muscle Fat Content in Boys With Duchenne Muscular Dystrophy. AJR Am J Roentgenol 2019; 212:1354-1360. [PMID: 30860898 DOI: 10.2214/ajr.18.20354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE. The purpose of this study was to investigate the potential of high-speed T2-corrected multiecho (HISTO) MR spectroscopy (MRS) for rapidly quantifying the fat content of thigh muscles in children with Duchenne muscular dystrophy (DMD). SUBJECTS AND METHODS. This study prospectively enrolled 58 boys with DMD (mean age, 7.5 years; range, 4-11 years) and 30 age-matched healthy boys (mean age, 7.2 years; range, 4-11 years) at one institution over a 1-year period. T1- and T2-weighted, multiecho Dixon, and HISTO sequences were performed on the right adductor magnus and vastus lateralis muscles. The fat fractions of these muscles were acquired from HISTO and multiecho Dixon images. An experienced radiologist graded the degree of fat infiltration of the adductor magnus and vastus lateralis muscles on axial T1-weighted images. The Bland-Altman method was used to assess the consistency and repeatability of the HISTO sequence. Pearson linear correlation analysis was used to determine the correlation coefficient relating HISTO fat fraction to multiecho Dixon fat fraction values. Spearman rank correlation analysis was used to assess the relation between the HISTO fat fraction values and T1-weighted image fat infiltration grades. The independent t test was used to compare the HISTO fat fraction values of the boys with DMD with those of the healthy control subjects. RESULTS. Bland-Altman analysis showed that 95.5% of the HISTO fat fraction values of the adductor magnus were within the 95% CI. HISTO fat fraction and multiecho Dixon fat fraction values of the adductor magnus and vastus lateralis muscles were highly positively correlated (adductor magnus, r = 0.983; vastus lateralis, r = 0.967; p < 0.0001). HISTO fat fraction values were also highly positively correlated with the grades of fat infiltration on T1-weighted images (adductor magnus, r = 0.911; vastus lateralis, r = 0.937; p < 0.0001). The HISTO fat fraction of the adductor magnus muscle was 33.3% ± 22.6% and of the vastus lateralis muscle was 25.6% ± 20.3% in patients with DMD. The corresponding values were 2.9% ± 2.1% and 2.3% ± 1.9% in the control group. The differences were statistically significant (p < 0.0001). CONCLUSION. The HISTO sequence is a rapid and feasible noninvasive MRS technique for quantifying the fat infiltration of thigh muscles in children with known or suspected DMD. It is useful for diagnosis and for assessment of disease activity and prognosis.
Collapse
|
18
|
Tomas X, Milisenda JC, Garcia-Diez AI, Prieto-Gonzalez S, Faruch M, Pomes J, Grau-Junyent JM. Whole-body MRI and pathological findings in adult patients with myopathies. Skeletal Radiol 2019; 48:653-676. [PMID: 30377729 DOI: 10.1007/s00256-018-3107-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/12/2018] [Accepted: 10/22/2018] [Indexed: 02/08/2023]
Abstract
Magnetic resonance imaging (MRI) is considered the most sensitive and specific imaging technique for the detection of muscle diseases related to myopathies. Since 2008, the use of whole-body MRI (WBMRI) to evaluate myopathies has improved due to technical advances such as rolling table platform and parallel imaging, which enable rapid assessment of the entire musculoskeletal system with high-quality images. WBMRI protocols should include T1-weighted and short-tau inversion recovery (STIR), which provide the basic pulse sequences for studying myopathies, in order to detect fatty infiltration/muscle atrophy and muscle edema, respectively. High signal intensity in T1-weighted images shows chronic disease with fatty infiltration, whereas high signal intensity in STIR indicates an acute stage with muscle edema. Additional sequences such as diffusion-weighted imaging (DWI) can be readily incorporated into routine WBMRI study protocols. Contrast-enhanced sequences have not been done. This article reviews WBMRI as an imaging method to evaluate different myopathies (idiopathic inflammatory, dystrophic, non-dystrophic, metabolic, and channelopathies). WBMRI provides a comprehensive estimate of the total burden with a single study, seeking specific distribution patterns, including clinically silent involvement of muscle areas. Furthermore, WBMRI may help to select the "target muscle area" for biopsy during patient follow-up. It may be also be used to detect related and non-related pathological conditions, such as tumors.
Collapse
Affiliation(s)
- Xavier Tomas
- Department of Radiology (CDIC), Hospital Clinic, Universitat de Barcelona (UB), Villarroel 170, 08036, Barcelona, Spain.
| | - Jose Cesar Milisenda
- Department of Internal Medicine, Hospital Clinic, Universitat de Barcelona (UB) and CIBERER, Villarroel 170, 08036, Barcelona, Spain
| | - Ana Isabel Garcia-Diez
- Department of Radiology (CDIC), Hospital Clinic, Universitat de Barcelona (UB), Villarroel 170, 08036, Barcelona, Spain
| | - Sergio Prieto-Gonzalez
- Department of Autoimmune Diseases, Hospital Clinic, Universitat de Barcelona (UB), Villarroel 170, 08036, Barcelona, Spain
| | - Marie Faruch
- Department of Radiology, Hopital Purpan, Centre Hospitalier Universitaire (CHU), Place du Docteur Baylac TSA 40031, 31059, Toulouse cedex 9, France
| | - Jaime Pomes
- Department of Radiology (CDIC), Hospital Clinic, Universitat de Barcelona (UB), Villarroel 170, 08036, Barcelona, Spain
| | - Josep Maria Grau-Junyent
- Department of Internal Medicine, Hospital Clinic, Universitat de Barcelona (UB) and CIBERER, Villarroel 170, 08036, Barcelona, Spain
| |
Collapse
|
19
|
Eresen A, Birch SM, Alic L, Griffin JF, Kornegay JN, Ji JX. New Similarity Metric for Registration of MRI to Histology: Golden Retriever Muscular Dystrophy Imaging. IEEE Trans Biomed Eng 2019; 66:1222-1230. [DOI: 10.1109/tbme.2018.2870711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
20
|
Cai J, Xing F, Batra A, Liu F, Walter GA, Vandenborne K, Yang L. Texture Analysis for Muscular Dystrophy Classification in MRI with Improved Class Activation Mapping. PATTERN RECOGNITION 2019; 86:368-375. [PMID: 31105339 PMCID: PMC6521874 DOI: 10.1016/j.patcog.2018.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The muscular dystrophies are made up of a diverse group of rare genetic diseases characterized by progressive loss of muscle strength and muscle damage. Since there is no cure for muscular dystrophy and clinical outcome measures are limited, it is critical to assess the progression of MD objectively. Imaging muscle replacement by fibrofatty tissue has been shown to be a robust biomarker to monitor disease progression in DMD. In magnetic resonance imaging (MRI) data, specific texture patterns are found to correlate to certain MD subtypes and thus present a potential way for automatic assessment. In this paper, we first apply state-of-the-art convolutional neural networks (CNNs) to perform accurate MD image classification and then propose an effective visualization method to highlight the important image textures. With a dystrophic MRI dataset, we found that the best CNN model delivers an 91.7% classification accuracy, which significantly outperforms non-deep learning methods, e.g., >40% improvement has been found over the traditional mean fat fraction (MFF) criterion for DMD and CMD classification. After investigating every single neuron at the top layer of CNN model, we found the superior classification ability of CNN can be explained by its 91 and 118 neurons were performing better than the MFF criterion under the measurements of Euclidean and Chi-square distance, respectively. In order to further interpret CNNs predictions, we tested an improved class activation mapping (ICAM) method to visualize the important regions in the MRI images. With this ICAM, CNNs are able to locate the most discriminative texture patterns of DMD in soleus, lateral gastrocnemius, and medial gastrocnemius; for CMD, the critical texture patterns are highlighted in soleus, tibialis posterior, and peroneus.
Collapse
Affiliation(s)
- Jinzheng Cai
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida
| | - Fuyong Xing
- Department of Biostatistics and Informatics, University of Colorado Denver
| | - Abhinandan Batra
- Department of Physiology and Functional Genomics, University of Florida
| | - Fujun Liu
- Department of Electrical and Computer Engineering, University of Florida
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida
| | | | - Lin Yang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida
- Department of Electrical and Computer Engineering, University of Florida
| |
Collapse
|
21
|
Kapur K, Sanchez B, Pacheck A, Darras B, Rutkove SB, Selukar R. Functional Mixed-Effects Modeling of Longitudinal Duchenne Muscular Dystrophy Electrical Impedance Myography Data Using State-Space Approach. IEEE Trans Biomed Eng 2018; 66:1761-1768. [PMID: 30387720 DOI: 10.1109/tbme.2018.2879227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Electrical impedance myography (EIM) is a quantitative and objective tool to evaluate muscle status. EIM offers the possibility to replace conventional physical functioning scores or quality of life measures, which depend on patient cooperation and mood. METHODS Here, we propose a functional mixed-effects model using a state-space approach to describe the response trajectories of EIM data measured on 16 boys with Duchenne muscular dystrophy and 12 healthy controls, both groups measured over a period of two years. The modeling framework presented imposes a smoothing spline structure on EIM data collected at each visit and taking into account of within subject correlations of these curves along the longitudinal measurements. The modeling framework is recast in a state-space approach, thereby allowing for the employment of computationally efficient diffuse Kalman filtering and smoothing algorithms for the model estimation, as well as the estimates of the posterior variance-covariance matrix for the construction of the Bayesian [Formula: see text] confidence bands. RESULTS The proposed model allows us to simultaneously adjust for baseline variables, differentiate the longitudinal changes in the smooth functional response and estimate the subject and subject-time specific deviations from the population-averaged response curves. The code is made publicly available in the supplementary material. SIGNIFICANCE The modeling approach presented will potentially enhance EIM capability to serve as a biomarker for testing therapeutic efficacy in DMD and other clinical trials.
Collapse
|
22
|
Park JS, Vohra R, Klussmann T, Bengtsson NE, Chamberlain JS, Lee D. Non-invasive tracking of disease progression in young dystrophic muscles using multi-parametric MRI at 14T. PLoS One 2018; 13:e0206323. [PMID: 30365532 PMCID: PMC6203357 DOI: 10.1371/journal.pone.0206323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 10/10/2018] [Indexed: 12/02/2022] Open
Abstract
In this study, multi-parametric magnetic resonance imaging (MRI) was conducted to monitor skeletal muscle changes in dystrophic (mdx4cv) and age-matched control (C57BL/6J) mice starting at 3 weeks of age. The objective of this study was to evaluate and characterize changes in muscle tissue characteristics of hind limbs in young, dystrophic mice using MRI. Mdx4cv (n = 25) and age-matched C57BL/6J (n = 5) were imaged at 3, 5, 7, 9, and 11 weeks of age. Multiple MR measurements were taken from the tibialis anterior, gastrocnemius, and soleus muscles. There were significant differences between dystrophic and control groups for all three muscle types when comparing transverse relaxation times (T2) in lower hind limb muscles. Additionally, fractional anisotropy, radial diffusivity, and eigenvalue analysis of diffusion tensor imaging also demonstrated significant differences between groups. Longitudinal relaxation times (T1) displayed no significant differences between groups. The earliest time points in the magnetization transfer ratio measurements displayed a significant difference. Histological analysis revealed significant differences in the tibialis anterior and gastrocnemius muscles between groups with the mdx mice displaying greater variability in muscle fiber size in later time points. The multi-parametric MRI approach offers a promising alternative for future development of a noninvasive avenue for tracking both disease progression and treatment response.
Collapse
Affiliation(s)
- Joshua S. Park
- Department of Radiology, University of Washington, Seattle, WA, United States of America
| | - Ravneet Vohra
- Department of Radiology, University of Washington, Seattle, WA, United States of America
| | - Thomas Klussmann
- Department of Radiology, University of Washington, Seattle, WA, United States of America
| | - Niclas E. Bengtsson
- Department of Neurology, University of Washington, Seattle, WA, United States of America
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, WA, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, WA, United States of America
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, WA, United States of America
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Donghoon Lee
- Department of Radiology, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
23
|
Chrzanowski SM, Vohra RS, Lee-McMullen BA, Batra A, Spradlin RA, Morales J, Forbes S, Vandenborne K, Barton ER, Walter GA. Contrast-Enhanced Near-Infrared Optical Imaging Detects Exacerbation and Amelioration of Murine Muscular Dystrophy. Mol Imaging 2018; 16:1536012117732439. [PMID: 29271299 PMCID: PMC5985549 DOI: 10.1177/1536012117732439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Assessment of muscle pathology is a key outcome measure to measure the success of
clinical trials studying muscular dystrophies; however, few robust minimally invasive
measures exist. Indocyanine green (ICG)-enhanced near-infrared (NIR) optical imaging
offers an objective, minimally invasive, and longitudinal modality that can quantify
pathology within muscle by imaging uptake of ICG into the damaged muscles. Dystrophic mice
lacking dystrophin (mdx) or gamma-sarcoglycan (Sgcg−/−) were compared to
control mice by NIR optical imaging and magnetic resonance imaging (MRI). We determined
that optical imaging could be used to differentiate control and dystrophic mice, visualize
eccentric muscle induced by downhill treadmill running, and restore the membrane integrity
in Sgcg−/− mice following adeno-associated virus (AAV) delivery of recombinant
human SGCG (desAAV8hSGCG). We conclude that NIR optical imaging is comparable to MRI and
can be used to detect muscle damage in dystrophic muscle as compared to unaffected
controls, monitor worsening of muscle pathology in muscular dystrophy, and assess
regression of pathology following therapeutic intervention in muscular dystrophies.
Collapse
Affiliation(s)
- Stephen M Chrzanowski
- 1 Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Ravneet S Vohra
- 1 Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | | | - Abhinandan Batra
- 3 Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Ray A Spradlin
- 4 Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Jazmine Morales
- 4 Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Sean Forbes
- 3 Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Krista Vandenborne
- 3 Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Elisabeth R Barton
- 4 Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Glenn A Walter
- 1 Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
24
|
Abstract
The major forms of autoimmune myopathies include dermatomyositis (DM), polymyositis (PM), myositis associated with antisynthetase syndrome (ASS), immune-mediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM). While each of these conditions has unique clinical and histopathological features, they all share an immune-mediated component. These conditions can occur in isolation or can be associated with systemic malignancies or connective tissue disorders (overlap syndromes). As more has been learned about these conditions, it has become clear that traditional classification schemes do not adequately group patients according to shared clinical features and prognosis. Newer classifications are now utilizing myositis-specific autoantibodies which correlate with clinical and histopathological phenotypes and risk of malignancy, and help in offering prognostic information with regard to treatment response. Based on observational data and expert opinion, corticosteroids are considered first-line therapy for DM, PM, ASS, and IMNM, although intravenous immunoglobulin (IVIG) is increasingly being used as initial therapy in IMNM related to statin use. Second-line agents are often required, but further prospective investigation is required regarding the optimal choice and timing of these agents.
Collapse
Affiliation(s)
- Emer R McGrath
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA.
| | - Christopher T Doughty
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Anthony A Amato
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| |
Collapse
|
25
|
Barnard AM, Willcocks RJ, Finanger EL, Daniels MJ, Triplett WT, Rooney WD, Lott DJ, Forbes SC, Wang DJ, Senesac CR, Harrington AT, Finkel RS, Russman BS, Byrne BJ, Tennekoon GI, Walter GA, Sweeney HL, Vandenborne K. Skeletal muscle magnetic resonance biomarkers correlate with function and sentinel events in Duchenne muscular dystrophy. PLoS One 2018; 13:e0194283. [PMID: 29554116 PMCID: PMC5858773 DOI: 10.1371/journal.pone.0194283] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To provide evidence for quantitative magnetic resonance (qMR) biomarkers in Duchenne muscular dystrophy by investigating the relationship between qMR measures of lower extremity muscle pathology and functional endpoints in a large ambulatory cohort using a multicenter study design. METHODS MR spectroscopy and quantitative imaging were implemented to measure intramuscular fat fraction and the transverse magnetization relaxation time constant (T2) in lower extremity muscles of 136 participants with Duchenne muscular dystrophy. Measures were collected at 554 visits over 48 months at one of three imaging sites. Fat fraction was measured in the soleus and vastus lateralis using MR spectroscopy, while T2 was assessed using MRI in eight lower extremity muscles. Ambulatory function was measured using the 10m walk/run, climb four stairs, supine to stand, and six minute walk tests. RESULTS Significant correlations were found between all qMR and functional measures. Vastus lateralis qMR measures correlated most strongly to functional endpoints (|ρ| = 0.68-0.78), although measures in other rapidly progressing muscles including the biceps femoris (|ρ| = 0.63-0.73) and peroneals (|ρ| = 0.59-0.72) also showed strong correlations. Quantitative MR biomarkers were excellent indicators of loss of functional ability and correlated with qualitative measures of function. A VL FF of 0.40 was an approximate lower threshold of muscle pathology associated with loss of ambulation. DISCUSSION Lower extremity qMR biomarkers have a robust relationship to clinically meaningful measures of ambulatory function in Duchenne muscular dystrophy. These results provide strong supporting evidence for qMR biomarkers and set the stage for their potential use as surrogate outcomes in clinical trials.
Collapse
Affiliation(s)
- Alison M. Barnard
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Rebecca J. Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Erika L. Finanger
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Michael J. Daniels
- Department of Statistics, University of Florida, Gainesville, FL, United States of America
| | - William T. Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Donovan J. Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Dah-Jyuu Wang
- Department of Radiology, Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Claudia R. Senesac
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Ann T. Harrington
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | | | - Barry S. Russman
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Barry J. Byrne
- Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida, Gainesville, FL, United States of America
| | - Gihan I. Tennekoon
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States of America
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States of America
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
26
|
A data-oriented self-calibration and robust chemical-shift encoding by using clusterization (OSCAR): Theory, optimization and clinical validation in neuromuscular disorders. Magn Reson Imaging 2017; 45:84-96. [PMID: 28982632 DOI: 10.1016/j.mri.2017.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 12/15/2022]
Abstract
Multi-echo Chemical Shift-Encoded (CSE) methods for Fat-Water quantification are growing in clinical use due to their ability to estimate and correct some confounding effects. State of the art CSE water/fat separation approaches rely on a multi-peak fat spectrum with peak frequencies and relative amplitudes kept constant over the entire MRI dataset. However, the latter approximation introduces a systematic error in fat percentage quantification in patients where the differences in lipid chemical composition are significant (such as for neuromuscular disorders) because of the spatial dependence of the peak amplitudes. The present work aims to overcome this limitation by taking advantage of an unsupervised clusterization-based approach offering a reliable criterion to carry out a data-driven segmentation of the input MRI dataset into multiple regions. Results established that the presented algorithm is able to identify at least 4 different partitions from MRI dataset under which to perform independent self-calibration routines and was found robust in NMD imaging studies (as evaluated on a cohort of 24 subjects) against latest CSE techniques with either calibrated or non-calibrated approaches. Particularly, the PDFF of the thigh was more reproducible for the quantitative estimation of pathological muscular fat infiltrations, which may be promising to evaluate disease progression in clinical practice.
Collapse
|
27
|
|
28
|
Weng WC, Tsui PH, Lin CW, Lu CH, Lin CY, Shieh JY, Lu FL, Ee TW, Wu KW, Lee WT. Evaluation of muscular changes by ultrasound Nakagami imaging in Duchenne muscular dystrophy. Sci Rep 2017; 7:4429. [PMID: 28667314 PMCID: PMC5493629 DOI: 10.1038/s41598-017-04131-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common debilitating muscular disorder. Developing a noninvasive measure for monitoring the progression of this disease is critical. The present study tested the effectiveness of using ultrasound Nakagami imaging to evaluate the severity of the dystrophic process. A total of 47 participants (40 with DMD and 7 healthy controls) were recruited. Patients were classified into stage 1 (presymptomatic and ambulatory), stage 2 (early nonambulatory), and stage 3 (late nonambulatory). All participants underwent ultrasound examinations on the rectus femoris, tibialis anterior, and gastrocnemius. The results revealed that the ultrasound Nakagami parameter correlated positively with functional severity in the patients with DMD. The median Nakagami parameter of the gastrocnemius muscle increased from 0.50 to 0.85, corresponding to the largest dynamic range between normal and stage 3. The accuracy, sensitivity, and specificity of diagnosing walking function were 85.52%, 76.31%, and 94.73%, respectively. The Nakagami parameter of the rectus femoris and gastrocnemius muscles correlated negatively with the 6-minute walking distance in the ambulatory patients. Therefore, changes in the Nakagami parameter for the gastrocnemius muscle are suitable for monitoring disease progression in ambulatory patients and for predicting ambulation loss. Ultrasound Nakagami imaging shows potential for evaluating patients with DMD.
Collapse
Affiliation(s)
- Wen-Chin Weng
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Po-Hsiang Tsui
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute for Radiological Research, Chang Gung University and Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Wei Lin
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Chun-Hao Lu
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yen Lin
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pediatrics, Yong-He Cardinal Tien Hospital, Taipei, Taiwan
| | - Jeng-Yi Shieh
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Taipei, Taiwan
| | - Frank Leigh Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Ting-Wei Ee
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuan-Wen Wu
- Department of Orthopaedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Wang-Tso Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan. .,Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan. .,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
29
|
Muscle MRI in pediatrics: clinical, pathological and genetic correlation. Pediatr Radiol 2017; 47:724-735. [PMID: 28102454 DOI: 10.1007/s00247-016-3777-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/31/2016] [Accepted: 12/28/2016] [Indexed: 10/20/2022]
Abstract
Pediatric myopathies comprise a very heterogeneous group of disorders that may develop at different ages and affect different muscle groups. Its diagnosis is sometimes difficult and must be confirmed by muscle biopsy and/or genetic analysis. In recent years, muscle involvement patterns observed on MRI have become a valuable tool, aiding clinical diagnosis and enriching pathological and genetic assessments. We selected eight myopathy cases from our institutional database in which the pattern of muscle involvement observed on MRI was almost pathognomonic and could therefore contribute to establishing diagnosis. Muscle biopsy, genetic diagnosis or both confirmed all cases.
Collapse
|
30
|
Schmidt S, Gocheva V, Zumbrunn T, Rubino-Nacht D, Bonati U, Fischer D, Hafner P. Treatment with L-citrulline in patients with post-polio syndrome: study protocol for a single-center, randomised, placebo-controlled, double-blind trial. Trials 2017; 18:116. [PMID: 28274276 PMCID: PMC5343398 DOI: 10.1186/s13063-017-1829-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/08/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Post-polio syndrome (PPS) is a condition that affects polio survivors years after recovery from an initial acute infection by the Poliomyelitis virus. Most often, patients who suffered from polio start to experience gradual new weakening in muscles, a gradual decrease in the size of muscles (muscle atrophy) and fatigue years after the acute illness. L-citrulline is known to change muscular metabolism synthesis by raising nitric oxide (NO) levels and increasing protein synthesis. This investigator-initiated, randomised, placebo-controlled, double-blind, trial aims to demonstrate that L-citrulline positively influences muscle function and increases muscular energy production in patients with PPS. METHODS/DESIGN Thirty ambulant PPS patients will be recruited in Switzerland. Patients will be randomly allocated to one of the two arms of the study (placebo:verum 1:1). After a 24-week run-in phase to observe natural disease history and progression, participants will be treated either with L-citrulline or placebo for 24 weeks. The primary endpoint is change in the 6-min Walking Distance Test. Secondary endpoints will include motor function measure, quantitative muscle force, quantitative muscle magnetic resonance imaging and magnetic resonance spectroscopy and serum biomarker laboratory analysis DISCUSSION: The aim of this phase IIa trial is to determine if treatment with L-citrulline shows a positive effect on clinical function and paraclinical biomarkers in PPS. If treatment with L-citrulline shows positive effects, this might represent a cost-efficient symptomatic therapy for PPS patients. TRIAL REGISTRATION ClinicalTrial.gov, ID: NCT02801071 . Registered on 6 June 2016.
Collapse
Affiliation(s)
- Simone Schmidt
- Division of Neuropediatrics, University of Basel Children’s Hospital, Spitalstrasse 33, Postfach 4031 Basel, Switzerland
- Division of Neurology, University Hospital Basel, Basel, Switzerland
| | - Vanya Gocheva
- Division of Neuropediatrics, University of Basel Children’s Hospital, Spitalstrasse 33, Postfach 4031 Basel, Switzerland
| | - Thomas Zumbrunn
- Department of Clinical Research, Clinical Trial Unit, University Hospital Basel, Basel, Switzerland
| | - Daniela Rubino-Nacht
- Division of Neuropediatrics, University of Basel Children’s Hospital, Spitalstrasse 33, Postfach 4031 Basel, Switzerland
| | - Ulrike Bonati
- Division of Neuropediatrics, University of Basel Children’s Hospital, Spitalstrasse 33, Postfach 4031 Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Dirk Fischer
- Division of Neuropediatrics, University of Basel Children’s Hospital, Spitalstrasse 33, Postfach 4031 Basel, Switzerland
- Division of Neurology, University Hospital Basel, Basel, Switzerland
- Division of Neurology, Medical University Clinic, Kantonsspital Baselland, Bruderholz, Switzerland
| | - Patricia Hafner
- Division of Neuropediatrics, University of Basel Children’s Hospital, Spitalstrasse 33, Postfach 4031 Basel, Switzerland
- Division of Neurology, Medical University Clinic, Kantonsspital Baselland, Bruderholz, Switzerland
| |
Collapse
|
31
|
Correa-de-Araujo R, Harris-Love MO, Miljkovic I, Fragala MS, Anthony BW, Manini TM. The Need for Standardized Assessment of Muscle Quality in Skeletal Muscle Function Deficit and Other Aging-Related Muscle Dysfunctions: A Symposium Report. Front Physiol 2017; 8:87. [PMID: 28261109 PMCID: PMC5310167 DOI: 10.3389/fphys.2017.00087] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022] Open
Abstract
A growing body of scientific literature suggests that not only changes in skeletal muscle mass, but also other factors underpinning muscle quality, play a role in the decline in skeletal muscle function and impaired mobility associated with aging. A symposium on muscle quality and the need for standardized assessment was held on April 28, 2016 at the International Conference on Frailty and Sarcopenia Research in Philadelphia, Pennsylvania. The purpose of this symposium was to provide a venue for basic science and clinical researchers and expert clinicians to discuss muscle quality in the context of skeletal muscle function deficit and other aging-related muscle dysfunctions. The present article provides an expanded introduction concerning the emerging definitions of muscle quality and a potential framework for scientific inquiry within the field. Changes in muscle tissue composition, based on excessive levels of inter- and intra-muscular adipose tissue and intramyocellular lipids, have been found to adversely impact metabolism and peak force generation. However, methods to easily and rapidly assess muscle tissue composition in multiple clinical settings and with minimal patient burden are needed. Diagnostic ultrasound and other assessment methods continue to be developed for characterizing muscle pathology, and enhanced sonography using sensors to provide user feedback and improve reliability is currently the subject of ongoing investigation and development. In addition, measures of relative muscle force such as specific force or grip strength adjusted for body size have been proposed as methods to assess changes in muscle quality. Furthermore, performance-based assessments of muscle power via timed tests of function and body size estimates, are associated with lower extremity muscle strength may be responsive to age-related changes in muscle quality. Future aims include reaching consensus on the definition and standardized assessments of muscle quality, and providing recommendations to address critical clinical and technology research gaps within the field.
Collapse
Affiliation(s)
- Rosaly Correa-de-Araujo
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, National Institutes of Health, U.S. Department of Health and Human Services Bethesda, MD, USA
| | - Michael O Harris-Love
- Muscle Morphology, Mechanics and Performance Laboratory, Clinical Research Center - Human Performance Research Unit, Veterans Affairs Medical CenterWashington, DC, USA; Geriatrics and Extended Care Service/Research Service, Veterans Affairs Medical CenterWashington, DC, USA; Department of Exercise and Nutritional Sciences, Milken Institute School of Public Health, The George Washington UniversityWashington, DC, USA
| | - Iva Miljkovic
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh Pittsburgh, PA, USA
| | | | - Brian W Anthony
- Laboratory for Manufacturing and Productivity, Massachusetts Institute of TechnologyCambridge, MA, USA; Medical Electronic Device Realization Center, Massachusetts Institute of TechnologyCambridge, MA, USA
| | - Todd M Manini
- Department of Aging & Geriatric Research, Institute on Aging, University of Florida College of Medicine Gainesville, FL, USA
| |
Collapse
|
32
|
Goemans N, vanden Hauwe M, Signorovitch J, Swallow E, Song J. Individualized Prediction of Changes in 6-Minute Walk Distance for Patients with Duchenne Muscular Dystrophy. PLoS One 2016; 11:e0164684. [PMID: 27737016 PMCID: PMC5063281 DOI: 10.1371/journal.pone.0164684] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/29/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Deficits in ambulatory function progress at heterogeneous rates among individuals with Duchenne muscular dystrophy (DMD). The resulting inherent variability in ambulatory outcomes has complicated the design of drug efficacy trials and clouded the interpretation of trial results. We developed a prediction model for 1-year change in the six minute walk distance (6MWD) among DMD patients, and compared its predictive value to that of commonly used prognostic factors (age, baseline 6MWD, and steroid use). METHODS Natural history data were collected from DMD patients at routine follow up visits approximately every 6 months over the course of 2-5 years. Assessments included ambulatory function and steroid use. The annualized change in 6MWD (Δ6MWD) was studied between all pairs of visits separated by 8-16 months. Prediction models were developed using multivariable regression for repeated measures, and evaluated using cross-validation. RESULTS Among n = 191 follow-up intervals (n = 39 boys), mean starting age was 9.4 years, mean starting 6MWD was 351.8 meters, and 75% had received steroids for at least one year. Over the subsequent 8-16 months, mean Δ6MWD was -37.0 meters with a standard deviation (SD) of 93.7 meters. Predictions based on a composite of age, baseline 6MWD, and steroid use explained 28% of variation in Δ6MWD (R2 = 0.28, residual SD = 79.4 meters). A broadened prognostic model, adding timed 10-meter walk/run, 4-stair climb, and rise from supine, as well as height and weight, significantly improved prediction, explaining 59% of variation in Δ6MWD after cross-validation (R2 = 0.59, residual SD = 59.7 meters). CONCLUSIONS A prognostic model incorporating timed function tests significantly improved prediction of 1-year changes in 6MWD. Explained variation was more than doubled compared to predictions based only on age, baseline 6MWD, and steroid use. There is significant potential for composite prognostic models to inform DMD clinical trials and clinical practice.
Collapse
Affiliation(s)
| | | | - James Signorovitch
- Analysis Group, Inc., 111 Huntington Ave, 14 floor, Boston, Massachusetts, United States of America
- The Trajectory Analysis Project (TAP) Collaboration, One Broadway, 14 floor, Cambridge, Massachusetts, United States of America
| | - Elyse Swallow
- Analysis Group, Inc., 111 Huntington Ave, 14 floor, Boston, Massachusetts, United States of America
| | - Jinlin Song
- Analysis Group, Inc., 111 Huntington Ave, 14 floor, Boston, Massachusetts, United States of America
| | | |
Collapse
|
33
|
Siracusano G, La Corte A, Milazzo C, Anastasi GP, Finocchio G, Gaeta M. On the R 2⁎ relaxometry in complex multi-peak multi-Echo chemical shift-based water-fat quantification: Applications to the neuromuscular diseases. Magn Reson Imaging 2016; 35:4-14. [PMID: 27569370 DOI: 10.1016/j.mri.2016.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 08/03/2016] [Accepted: 08/20/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Investigation of the feasibility of the R2⁎ mapping techniques by using latest theoretical models corrected for confounding factors and optimized for signal to noise ratio. THEORY AND METHODS The improvement of the performance of state of the art magnetic resonance imaging (MRI) relaxometry algorithms is challenging because of a non-negligible bias and still unresolved numerical instabilities. Here, R2⁎ mapping reconstructions, including complex fitting with multi-spectral fat-correction by using single-decay and double-decay formulation, are deeply studied in order to investigate and identify optimal configuration parameters and minimize the occurrence of numerical artifacts. The effects of echo number, echo spacing, and fat/water relaxation model type are evaluated through both simulated and in-vivo data. We also explore the stability and feasibility of the fat/water relaxation model by analyzing the impact of high percentage of fat infiltrations and local transverse relaxation differences among biological species. RESULTS The main limits of the MRI relaxometry are the presence of bias and the occurrence of artifacts, which significantly affect its accuracy. Chemical-shift complex R2⁎-correct single-decay reconstructions exhibit a large bias in presence of a significant difference in the relaxation rates of fat and water and with fat concentration larger than 30%. We find that for fat-dominated tissues or in patients affected by extensive iron deposition, MRI reconstructions accounting for multi-exponential relaxation time provide accurate R2⁎ measurements and are less prone to numerical artifacts. CONCLUSIONS Complex fitting and fat-correction with multi-exponential decay formulation outperforms the conventional single-decay approximation in various diagnostic scenarios. Although it still lacks of numerical stability, which requires model enhancement and support from spectroscopy, it offers promising perspectives for the development of relaxometry as a reliable tool to improve tissue characterization and monitoring of neuromuscular disorders.
Collapse
Affiliation(s)
- Giulio Siracusano
- Department of Mathematical and Computer Sciences, Physical Sciences and Earth Sciences, University of Messina, V.le F. D'alcontres, 31, 98166, Messina, Italy; Department of Computer Engineering and Telecommunications, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy.
| | - Aurelio La Corte
- Department of Computer Engineering and Telecommunications, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Carmelo Milazzo
- Department of Biomedical sciences, Dental and of Morphological and Functional images, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy
| | - Giuseppe Pio Anastasi
- Department of Biomedical sciences, Dental and of Morphological and Functional images, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy
| | - Giovanni Finocchio
- Department of Mathematical and Computer Sciences, Physical Sciences and Earth Sciences, University of Messina, V.le F. D'alcontres, 31, 98166, Messina, Italy; Istituto Nazionale di Geofisica e Vulcanologia (INGV), Via Vigna Murata 605, 00143, Roma, Italy
| | - Michele Gaeta
- Department of Biomedical sciences, Dental and of Morphological and Functional images, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy
| |
Collapse
|
34
|
Pigula AJ, Wu JS, Gilbertson MW, Darras BT, Rutkove SB, Anthony BW. Force-controlled ultrasound to measure passive mechanical properties of muscle in Duchenne muscular dystrophy. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2016; 2016:2865-2868. [PMID: 28324973 DOI: 10.1109/embc.2016.7591327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The purpose of this study is to assess differences in skeletal muscle compressibility between patients with Duchenne muscular dystrophy (DMD) and normal subjects. The transverse passive mechanical properties of muscle, particularly those related to stiffness and elasticity, can be measured using force-controlled ultrasound. We acquired ultrasound videos of muscle compression under known pressures in the biceps and quadriceps in 23 boys with DMD and 20 age-matched healthy controls. We calculated the bulk linear spring constant, nonlinear stress-strain response, and average Young's modulus for each. Young's modulus was found to be significantly higher in the DMD population in both the biceps (normal: 33 ± 6 kPa, DMD: 45 ± 14, p <; .01) and quadriceps (normal: 42 ± 6, DMD: 58 ± 14, p <; .0001). Muscle compressibility measured by force-controlled ultrasound is an objective and robust technique to quantitatively monitor the effects of DMD and distinguish from normal subjects.
Collapse
|
35
|
Erlandson M, Lorbergs A, Mathur S, Cheung A. Muscle analysis using pQCT, DXA and MRI. Eur J Radiol 2016; 85:1505-11. [DOI: 10.1016/j.ejrad.2016.03.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/22/2022]
|
36
|
Straub V, Balabanov P, Bushby K, Ensini M, Goemans N, De Luca A, Pereda A, Hemmings R, Campion G, Kaye E, Arechavala-Gomeza V, Goyenvalle A, Niks E, Veldhuizen O, Furlong P, Stoyanova-Beninska V, Wood MJ, Johnson A, Mercuri E, Muntoni F, Sepodes B, Haas M, Vroom E, Aartsma-Rus A. Stakeholder cooperation to overcome challenges in orphan medicine development: the example of Duchenne muscular dystrophy. Lancet Neurol 2016; 15:882-890. [DOI: 10.1016/s1474-4422(16)30035-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/23/2016] [Accepted: 03/31/2016] [Indexed: 01/05/2023]
|
37
|
Peverelli L, Testolin S, Villa L, D'Amico A, Petrini S, Favero C, Magri F, Morandi L, Mora M, Mongini T, Bertini E, Sciacco M, Comi GP, Moggio M. Histologic muscular history in steroid-treated and untreated patients with Duchenne dystrophy. Neurology 2015; 85:1886-93. [PMID: 26497992 DOI: 10.1212/wnl.0000000000002147] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 07/16/2015] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is a lethal disease. The outcome measures used in numerous therapeutic trials include skeletal muscle biopsy. We studied the natural history of DMD from the standpoint of muscle histology with the aim of providing a reproducible tool for use in evaluating and comparing any histologic changes occurring in patients with DMD undergoing treatment and hence be able to determine how therapy modulates the histologic evolution of the disease. METHODS Three independent operators analyzed 56 muscle biopsies from 40 patients not treated with steroids, aged 1 to 10 years and 16 individuals treated with steroids, aged 7 to 10 years. We analyzed morphologic measures, normalized every measure for the average number of fibers observed for each year of age, and calculated intraclass correlation coefficients. RESULTS The average proportion of connective tissue in patients not treated with steroids was 16.98% from ages 1 to 6 years and 30% from ages 7 to 10 years (p < 0.0001). The average proportion in patients treated with steroids was 24.90%. Muscle fiber area mirrored that of connective tissue in both groups. CONCLUSIONS Having provided a reproducible tool for evaluation and comparison of histologic changes occurring in patients undergoing clinical trials, it was observed that at ages 6 to 7 years, fibrotic tissue rapidly peaks to 29.85%; this is a crucial moment when muscle tissue loses its self-regeneration ability, veering toward fibrotic degeneration. These data should be considered when deciding the most suitable time to begin therapy.
Collapse
Affiliation(s)
- Lorenzo Peverelli
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Silvia Testolin
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Luisa Villa
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Adele D'Amico
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Stefania Petrini
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Chiara Favero
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Francesca Magri
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Lucia Morandi
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Marina Mora
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Tiziana Mongini
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Enrico Bertini
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Monica Sciacco
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Giacomo P Comi
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Maurizio Moggio
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy.
| |
Collapse
|
38
|
Optimal classification for the diagnosis of duchenne muscular dystrophy images using support vector machines. Int J Comput Assist Radiol Surg 2015; 11:1755-63. [PMID: 26476638 DOI: 10.1007/s11548-015-1312-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/29/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND This study aimed to investigate the optimal support vector machines (SVM)-based classifier of duchenne muscular dystrophy (DMD) magnetic resonance imaging (MRI) images. METHODS T1-weighted (T1W) and T2-weighted (T2W) images of the 15 boys with DMD and 15 normal controls were obtained. Textural features of the images were extracted and wavelet decomposed, and then, principal features were selected. Scale transform was then performed for MRI images. Afterward, SVM-based classifiers of MRI images were analyzed based on the radical basis function and decomposition levels. The cost (C) parameter and kernel parameter [Formula: see text] were used for classification. Then, the optimal SVM-based classifier, expressed as [Formula: see text]), was identified by performance evaluation (sensitivity, specificity and accuracy). RESULTS Eight of 12 textural features were selected as principal features (eigenvalues [Formula: see text]). The 16 SVM-based classifiers were obtained using combination of (C, [Formula: see text]), and those with lower C and [Formula: see text] values showed higher performances, especially classifier of [Formula: see text]). The SVM-based classifiers of T1W images showed higher performance than T1W images at the same decomposition level. The T1W images in classifier of [Formula: see text]) at level 2 decomposition showed the highest performance of all, and its overall correct sensitivity, specificity, and accuracy reached 96.9, 97.3, and 97.1 %, respectively. CONCLUSION The T1W images in SVM-based classifier [Formula: see text] at level 2 decomposition showed the highest performance of all, demonstrating that it was the optimal classification for the diagnosis of DMD.
Collapse
|
39
|
Govindarajan R, Shepard KM, Jones LK. Diagnosis and treatment of limb-girdle and distal dystrophies: Payment policy perspectives. Neurol Clin Pract 2015; 5:454-459. [PMID: 29443172 DOI: 10.1212/cpj.0000000000000188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Limb-girdle muscular dystrophies (LGMDs) and distal dystrophies are a diverse group of genetically heterogeneous myopathies characterized by an evolving and often confusing nomenclature. Though rare as a group, they are commonly seen in neuromuscular clinics and occasionally in general neurology clinics, and are frequently a source of diagnostic dilemma. A recent evidence-based guideline by the American Academy of Neurology provides a comprehensive analysis of the clinical phenotypes, diagnostic approach, and management principles of the LGMDs and associated disorders. There remain many unanswered questions regarding the role of radiologic and genetic testing, cardiorespiratory screening, and physical therapy in managing these patients. This payment policy article suggests potential solutions to challenging coverage scenarios that result from incomplete or conflicting evidence.
Collapse
Affiliation(s)
- Raghav Govindarajan
- University of Missouri (RG), Columbia; American Academy of Neurology (KMS), Minneapolis; and Mayo Clinic (LKJ), Rochester, MN
| | - Katie M Shepard
- University of Missouri (RG), Columbia; American Academy of Neurology (KMS), Minneapolis; and Mayo Clinic (LKJ), Rochester, MN
| | - Lyell K Jones
- University of Missouri (RG), Columbia; American Academy of Neurology (KMS), Minneapolis; and Mayo Clinic (LKJ), Rochester, MN
| |
Collapse
|
40
|
Wary C, Azzabou N, Giraudeau C, Le Louër J, Montus M, Voit T, Servais L, Carlier P. Quantitative NMRI and NMRS identify augmented disease progression after loss of ambulation in forearms of boys with Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2015; 28:1150-1162. [PMID: 26215733 DOI: 10.1002/nbm.3352] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 04/27/2015] [Accepted: 06/03/2015] [Indexed: 05/27/2023]
Abstract
Quantitative NMRI and (31)P NMRS indices are reported in the forearms of 24 patients with Duchenne muscular dystrophy (DMD) (6-18 years, 14 non-ambulant) amenable to exon 53 skipping therapy and in 12 age-matched male controls (CONT). Examinations carried out at 3 T comprised multi-slice 17-echo measurements of muscle water T2 and heterogeneity, three-point Dixon imaging of fat fraction in flexor and extensor muscles (FLEX, EXT), and non-localised spectroscopy of phosphate metabolites. We studied four imaging indices, eight metabolic ratios combining ATP, phosphocreatine, phosphomonoesters and phosphodiesters, the cytosolic inorganic phosphate (Pia ) and an alkaline (Pib) pool present in dystrophic muscle, and average pH. All indices differed between DMD and CONT, except for muscle water T2 . Measurements were outside the 95th percentile of age-matched CONT values in over 65% of cases for percentage fat signal (%F), and in 78-100% of cases for all spectroscopic indices. T2 was elevated in one-third of FLEX measurements, whereas %pixels > 39 ms and T2 heterogeneity were abnormal in one-half of the examinations. The FLEX muscles had higher fat infiltration and T2 than EXT muscle groups. All indices, except pH, correlated with patient age, although the correlation was negative for T2 . However, in non-ambulant patients, the correlation with years since loss of ambulation was stronger than the correlation with age, and the slope of evolution per year was steeper after loss of ambulation. All indices except Pi/gATP differed between ambulant and non-ambulant patients; however, T2 and %pixels > 39 ms were highest in ambulant patients, possibly owing to the greater extent of inflammatory processes earlier in the disease. All other indices were worse in non-ambulant subjects. Quantitative measurements obtained from patients at different disease stages covered a broad range of abnormalities that evolved with the disease, and metabolic indices were up to 10-fold above normal from the onset, thus establishing a variety of potential markers for future therapy.
Collapse
Affiliation(s)
- Claire Wary
- AIM-CEA, Institute of Myology, NMR Laboratory, Paris, France
- CEA, I2BM, MIRCen, IdM NMR Laboratory, Paris, France
- UPMC University, Paris 06, Paris, France
| | - Noura Azzabou
- AIM-CEA, Institute of Myology, NMR Laboratory, Paris, France
- CEA, I2BM, MIRCen, IdM NMR Laboratory, Paris, France
- UPMC University, Paris 06, Paris, France
| | - Céline Giraudeau
- AIM-CEA, Institute of Myology, NMR Laboratory, Paris, France
- CEA, I2BM, MIRCen, IdM NMR Laboratory, Paris, France
- UPMC University, Paris 06, Paris, France
| | - Julien Le Louër
- AIM-CEA, Institute of Myology, NMR Laboratory, Paris, France
- CEA, I2BM, MIRCen, IdM NMR Laboratory, Paris, France
- UPMC University, Paris 06, Paris, France
| | | | - Thomas Voit
- Institute of Myology, UPMC-INSERM U974, CNRS FRE 3617, Paris, France
| | - Laurent Servais
- Institute of Myology, Clinical Trial and Database Unit, Paris, France
| | - Pierre Carlier
- AIM-CEA, Institute of Myology, NMR Laboratory, Paris, France
- CEA, I2BM, MIRCen, IdM NMR Laboratory, Paris, France
- UPMC University, Paris 06, Paris, France
| |
Collapse
|
41
|
Li W, Zheng Y, Zhang W, Wang Z, Xiao J, Yuan Y. Progression and variation of fatty infiltration of the thigh muscles in Duchenne muscular dystrophy, a muscle magnetic resonance imaging study. Neuromuscul Disord 2015; 25:375-80. [DOI: 10.1016/j.nmd.2015.01.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 01/11/2015] [Accepted: 01/14/2015] [Indexed: 02/04/2023]
|
42
|
Park J, Wicki J, Knoblaugh SE, Chamberlain JS, Lee D. Multi-parametric MRI at 14T for muscular dystrophy mice treated with AAV vector-mediated gene therapy. PLoS One 2015; 10:e0124914. [PMID: 25856443 PMCID: PMC4391935 DOI: 10.1371/journal.pone.0124914] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/11/2015] [Indexed: 01/22/2023] Open
Abstract
The objective of this study was to investigate the efficacy of using quantitative magnetic resonance imaging (MRI) as a non-invasive tool for the monitoring of gene therapy for muscular dystrophy. The clinical investigations for this family of diseases often involve surgical biopsy which limits the amount of information that can be obtained due to the invasive nature of the procedure. Thus, other non-invasive tools may provide more opportunities for disease assessment and treatment responses. In order to explore this, dystrophic mdx4cv mice were systemically treated with a recombinant adeno-associated viral (AAV) vector containing a codon-optimized micro-dystrophin gene. Multi-parametric MRI of T2, magnetization transfer, and diffusion effects alongside 3-D volume measurements were then utilized to monitor disease/treatment progression. Mice were imaged at 10 weeks of age for pre-treatment, then again post-treatment at 8, 16, and 24 week time points. The efficacy of treatment was assessed by physiological assays for improvements in function and quantification of expression. Tissues from the hindlimbs were collected for histological analysis after the final time point for comparison with MRI results. We found that introduction of the micro-dystrophin gene restored some aspects of normal muscle histology and pathology such as decreased necrosis and resistance to contraction-induced injury. T2 relaxation values showed percentage decreases across all muscle types measured (tibialis anterior, gastrocnemius, and soleus) when treated groups were compared to untreated groups. Additionally, the differences between groups were statistically significant for the tibialis anterior as well. The diffusion measurements showed a wider range of percentage changes and less statistical significance while the magnetization transfer effect measurements showed minimal change. MR images displayed hyper-intense regions of muscle that correlated with muscle pathology in histological sections. T2 relaxation, alongside diffusion and magnetization transfer effects provides useful data towards the goal of non-invasively monitoring the treatment of muscular dystrophy.
Collapse
Affiliation(s)
- Joshua Park
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
| | - Jacqueline Wicki
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Sue E. Knoblaugh
- Comparative Medicine Shared Resources, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Donghoon Lee
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
43
|
Effectiveness of diffusion tensor imaging in assessing disease severity in Duchenne muscular dystrophy: preliminary study. Pediatr Radiol 2015; 45:582-9. [PMID: 25246097 DOI: 10.1007/s00247-014-3187-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/22/2014] [Accepted: 09/11/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND There is currently a lack of suitable objective endpoints to measure disease progression in Duchenne muscular dystrophy (DMD). Emerging research suggests that diffusion tensor imaging (DTI) has potential as an outcome measure for the evaluation of skeletal muscle injury. OBJECTIVE The objective of this study was to evaluate the potential of DTI as quantitative magnetic resonance imaging (MRI) markers of disease severity in DMD. MATERIALS AND METHODS Thirteen consecutive boys (8.9 years ± 3.0 years) with DMD were evaluated using DTI. Fractional anisotropy (FA) and apparent diffusion coefficient (ADC) were compared with clinical outcome measures of manual muscle testing and MRI determinations of muscle fat fraction (MFF) in the right lower extremity. RESULTS Both MRI measures of FA and ADC strongly correlated with age and muscle strength. Values for FA positively correlated with age and negatively correlated with muscle strength (r = 0.78 and -0.96; both P ≤ 0.002) while measures of ADC negatively correlated age, but positively correlated with muscle strength (r = -0.87 and 0.83; both P ≤ 0.0004). Additionally, ADC and FA strongly correlated with MFF (r = -0.891 and 0.894, respectively; both P ≤ 0.0001). Mean MMF was negatively correlated with muscle strength (r = -0.89, P = 0.0001). CONCLUSION DTI measures of muscle structure strongly correlated with muscle strength and adiposity in boys with DMD in this pilot study, although these markers may be more reflective of fat replacement rather than muscle damage in later stages of the disease. Further studies in presymptomatic younger children are needed to assess the ability of DTI to detect early changes in DMD.
Collapse
|
44
|
Leung DG, Carrino JA, Wagner KR, Jacobs MA. Whole-body magnetic resonance imaging evaluation of facioscapulohumeral muscular dystrophy. Muscle Nerve 2015; 52:512-20. [PMID: 25641525 DOI: 10.1002/mus.24569] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 12/22/2014] [Accepted: 01/07/2015] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Facioscapulohumeral muscular dystrophy (FSHD) is a hereditary disorder that causes progressive muscle wasting. Increasing knowledge of the pathophysiology of FSHD has stimulated interest in developing biomarkers of disease severity. METHODS Two groups of MRI scans were analyzed: whole-body scans from 13 subjects with FSHD; and upper and lower extremity scans from 34 subjects with FSHD who participated in the MYO-029 clinical trial. Muscles were scored for fat infiltration and edema-like changes. Fat infiltration scores were compared with muscle strength and function. RESULTS The analysis revealed a distinctive pattern of both frequent muscle involvement and frequent sparing in FSHD. Averaged fat infiltration scores for muscle groups in the legs correlated with quantitative muscle strength and 10-meter walk times. CONCLUSIONS Advances in MRI technology allow for acquisition of rapid, high-quality, whole-body imaging in diffuse muscle disease. This technique offers a promising disease biomarker in FSHD and other muscle diseases.
Collapse
Affiliation(s)
- Doris G Leung
- Center for Genetic Muscle Disorders, The Hugo W. Moser Research Institute, Kennedy Krieger Institute, 707 North Broadway, 400A, Baltimore, Maryland, 21205, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John A Carrino
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiology and Imaging, Hospital for Special Surgery, New York, New York, USA
| | - Kathryn R Wagner
- Center for Genetic Muscle Disorders, The Hugo W. Moser Research Institute, Kennedy Krieger Institute, 707 North Broadway, 400A, Baltimore, Maryland, 21205, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Jacobs
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Shklyar I, Pasternak A, Kapur K, Darras BT, Rutkove SB. Composite biomarkers for assessing Duchenne muscular dystrophy: an initial assessment. Pediatr Neurol 2015; 52:202-5. [PMID: 25447928 PMCID: PMC4336219 DOI: 10.1016/j.pediatrneurol.2014.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/08/2014] [Accepted: 09/20/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Compared with individual parameters, composite biomarkers may provide a more effective means for monitoring disease progression and the effects of therapy in clinical trials than single measures. In this study, we built composite biomarkers for use in Duchenne muscular dystrophy by combining values from two objective measures of disease severity: electrical impedance myography and quantitative ultrasound and evaluating how well they correlated to standard functional measures. METHODS Using data from an ongoing study of electrical impedance myography and quantitative ultrasound in 31 Duchenne muscular dystrophy and 26 healthy boys aged 2-14 years, we combined data sets by first creating z scores based on the normal subject data and then using simple mathematical operations (addition and multiplication) to create composite measures. These composite scores were then correlated to age and standard measures of function including the 6-minute walk test, the North Star Ambulatory Assessment, and handheld dynamometry. RESULTS Combining data sets resulted in stronger correlations with all four outcomes than for either electrical impedance myography or quantitative ultrasound alone in six of eight instances. These improvements reached statistical significance (P < 0.05) in several cases. For example, the correlation coefficient for the composite measure with the North Star Ambulatory Assessment was 0.79 but was only 0.66 and 0.67 (respectively) for gray scale level and electrical impedance myography separately. CONCLUSIONS Arithmetically derived composite scores can provide stronger correlations to functional measures than isolated biomarkers. Longitudinal study of such composite markers in Duchenne muscular dystrophy clinical trials is warranted.
Collapse
Affiliation(s)
- Irina Shklyar
- Departments of Neurology, Beth Israel Deaconess Medical Center,
Boston, MA
| | - Amy Pasternak
- Boston Children’s Hospital, Harvard Medical School, Boston,
MA
| | - Kush Kapur
- Boston Children’s Hospital, Harvard Medical School, Boston,
MA
| | - Basil T. Darras
- Boston Children’s Hospital, Harvard Medical School, Boston,
MA
| | - Seward B. Rutkove
- Departments of Neurology, Beth Israel Deaconess Medical Center,
Boston, MA
| |
Collapse
|
46
|
Li P, Cui K, Zhang B, Wang Z, Shen Y, Wang X, Zhang J, Tong F, Li S. Transplantation of human umbilical cord-derived mesenchymal stems cells for the treatment of Becker muscular dystrophy in affected pedigree members. Int J Mol Med 2015; 35:1051-7. [PMID: 25647308 DOI: 10.3892/ijmm.2015.2084] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/20/2015] [Indexed: 11/05/2022] Open
Abstract
The regeneration of muscle tissue has been achieved using multipotent mesenchymal stem cells in mouse models of injured skeletal muscle. In the present study, the utility of multipotent human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in the treatment of Becker muscular dystrophy (BMD), a genetic disease where muscle tissue fails to regenerate, was examined in members from a pedigree affected by BMD. The disease status was evaluated in 4 affected pedigree members (II1, II2, II3 and III2; aged 50, 46, 42 and 6 years, respectively). The transplantation of the hUC‑MSCs (performed on 3 patients, I2, II3 and III2) was performed by infusion with an intravenous drip over a 30‑min period, and the patients were evaluated at 1, 3, 4 and 12 weeks following the procedure. The evaluation was based on physical characteristics, as well as on molecular testing for serum creatine kinase (CK) and lactate dehydrogenase (LDH) levels and a histological examination of muscle biopsies. The patients suffered no adverse reactions in response to the transplantation of the hUC‑MSCs. At 1 week following transplantation all 3 patients showed improvement in the muscle force of the limbs, muscle size and daily activity. The walking gait of patient III2 had improved by 1 week post-transplantation and reached a normal status by 12 weeks. Serum CK and LDH levels were decreased relative to the baseline levels. A histological examination of muscle biopsies displayed no obvious tissue regeneration. In conclusion, the treatment of patients with BMD using hUC-MSCs was safe and of therapeutic benefit that lasted for up to 12 weeks. hUC-MSCs are, therefore, a potential cell therapy-based treatment option for patients with muscular dystrophies.
Collapse
Affiliation(s)
- Pang Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Kai Cui
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Bo Zhang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Zhendan Wang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Yangyang Shen
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Xiangyu Wang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Jianbo Zhang
- Department of Pathology, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Feng Tong
- The Dean's Office, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| | - Sheng Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Jinan, Shandong 250011, P.R. China
| |
Collapse
|
47
|
Díaz-Manera J, Llauger J, Gallardo E, Illa I. Muscle MRI in muscular dystrophies. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2015; 34:95-108. [PMID: 27199536 PMCID: PMC4859076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Muscle MRI has become a very useful tool in the diagnosis and follow-up of patients with muscle dystrophies. Muscle MRI provides us about many aspects of the structure and function of skeletal muscles, such as the presence of oedema or fatty infiltration. In the last years many reports have described the particular muscles that are involved in these muscle disease. This knowledge can facilitate the diagnosis in many cases. In the present paper we review the main changes observed in muscle MRI of patients with muscle dystrophies.
Collapse
Affiliation(s)
- Jordi Díaz-Manera
- Neuromuscular Disorders Unit, Neurology Department, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona;,Centro de Investigación Básica en Enfermedades Raras (CIBERER);,Address for correspondence: Dr. Jordi Díaz-Manera, Neurology Department, Hospital de la Santa Creu I Sant Pau de Barcelona, C/ Sant Antoni Mª Claret 167, 08025 Barcelona, Spain. E-mail:
| | - Jaume Llauger
- Radiology Department, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona
| | - Eduard Gallardo
- Neuromuscular Disorders Unit, Neurology Department, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona;,Centro de Investigación Básica en Enfermedades Raras (CIBERER)
| | - Isabel Illa
- Neuromuscular Disorders Unit, Neurology Department, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona;,Centro de Investigación Básica en Enfermedades Raras (CIBERER)
| |
Collapse
|
48
|
Lynn S, Aartsma-Rus A, Bushby K, Furlong P, Goemans N, De Luca A, Mayhew A, McDonald C, Mercuri E, Muntoni F, Pohlschmidt M, Verschuuren J, Voit T, Vroom E, Wells DJ, Straub V. Measuring clinical effectiveness of medicinal products for the treatment of Duchenne muscular dystrophy. Neuromuscul Disord 2015; 25:96-105. [DOI: 10.1016/j.nmd.2014.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/01/2014] [Indexed: 10/24/2022]
|
49
|
Beltran E, Shelton GD, Guo LT, Dennis R, Sanchez-Masian D, Robinson D, De Risio L. Dystrophin-deficient muscular dystrophy in a Norfolk terrier. J Small Anim Pract 2014; 56:351-4. [PMID: 25353637 DOI: 10.1111/jsap.12292] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/07/2014] [Accepted: 08/29/2014] [Indexed: 11/29/2022]
Abstract
A six-month-old male entire Norfolk terrier was presented with a 3-month history of poor development, reluctance to exercise and progressive and diffuse muscle atrophy. Serum creatine kinase concentration was markedly elevated. Magnetic resonance imaging of the epaxial muscles revealed asymmetrical streaky signal changes aligned within the muscle fibres (hyperintense on T2-weighted images and short-tau inversion recovery with moderate contrast enhancement on T1-weighted images). Electromyography revealed pseudomyotonic discharges and fibrillation potentials localised at the level of the supraspinatus, epaxial muscles and tibial cranialis muscles. Muscle biopsy results were consistent with dystrophin-deficient muscular dystrophy. The dog remained stable 7 months after diagnosis with coenzyme Q10 and l-carnitine; however after that time, there was a marked deterioration and the owners elected euthanasia. This case report describes the clinical presentation, magnetic resonance imaging, electrodiagnostic and histopathological findings with immunohistochemical analysis in a Norfolk terrier with confirmed dystrophin-deficient muscular dystrophy, which has not been previously described in this breed.
Collapse
Affiliation(s)
- E Beltran
- Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, CB87UU
| | | | | | | | | | | | | |
Collapse
|
50
|
Influence of a two-year steroid treatment on body composition as measured by dual X-ray absorptiometry in boys with Duchenne muscular dystrophy. Neuromuscul Disord 2014; 24:467-73. [DOI: 10.1016/j.nmd.2014.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 02/25/2014] [Accepted: 03/01/2014] [Indexed: 11/18/2022]
|