1
|
Saad FA, Siciliano G, Angelini C. Advances in Dystrophinopathy Diagnosis and Therapy. Biomolecules 2023; 13:1319. [PMID: 37759719 PMCID: PMC10526396 DOI: 10.3390/biom13091319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Dystrophinopathies are x-linked muscular disorders which emerge from mutations in the Dystrophin gene, including Duchenne and Becker muscular dystrophy, and dilated cardiomyopathy. However, Duchenne muscular dystrophy interconnects with bone loss and osteoporosis, which are exacerbated by glucocorticoids therapy. Procedures for diagnosing dystrophinopathies include creatine kinase assay, haplotype analysis, Southern blot analysis, immunological analysis, multiplex PCR, multiplex ligation-dependent probe amplification, Sanger DNA sequencing, and next generation DNA sequencing. Pharmacological therapy for dystrophinopathies comprises glucocorticoids (prednisone, prednisolone, and deflazacort), vamorolone, and ataluren. However, angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), and β-blockers are the first-line to prevent dilated cardiomyopathy in dystrophinopathy patients. Duchenne muscular dystrophy gene therapy strategies involve gene transfer, exon skipping, exon reframing, and CRISPR gene editing. Eteplirsen, an antisense-oligonucleotide drug for skipping exon 51 from the Dystrophin gene, is available on the market, which may help up to 14% of Duchenne muscular dystrophy patients. There are various FDA-approved exon skipping drugs including ExonDys-51 for exon 51, VyonDys-53 and Viltolarsen for exon 53 and AmonDys-45 for exon 45 skipping. Other antisense oligonucleotide drugs in the pipeline include casimersen for exon 45, suvodirsen for exon 51, and golodirsen for exon 53 skipping. Advances in the diagnosis and therapy of dystrophinopathies offer new perspectives for their early discovery and care.
Collapse
Affiliation(s)
- Fawzy A. Saad
- Department of Gene Therapy, Saad Pharmaceuticals, Juhkentali 8, 10132 Tallinn, Estonia
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, Pisa University School of Medicine, Via Paradisa 2, 56100 Pisa, Italy;
| | - Corrado Angelini
- Department of Neurosciences, Padova University School of Medicine, Via Giustiniani 5, 35128 Padova, Italy;
| |
Collapse
|
2
|
Tsygankova P, Bychkov I, Minzhenkova M, Pechatnikova N, Bessonova L, Buyanova G, Naumchik I, Beskorovainiy N, Tabakov V, Itkis Y, Shilova N, Zakharova E. Expanding the genetic spectrum of the pyruvate carboxylase deficiency with novel missense, deep intronic and structural variants. Mol Genet Metab Rep 2022; 32:100889. [PMID: 35782291 PMCID: PMC9240867 DOI: 10.1016/j.ymgmr.2022.100889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Pathogenic variants in the pyruvate carboxylase (PC) gene cause a wide spectrum of recessive phenotypes, ranging from the early-onset fatal encephalopathy to the adult-onset benign form. Results Patient 1 is a 6 y.o. boy with ataxia, hypoglycemia and episodes of lactic acidosis. WGS revealed the novel heterozygous missense variant c.1372A > G (p.Asn458Asp) in the PC gene. Additional analysis revealed discordant reads mapped to chromosomes 11 and 1, so a reciprocal translocation disrupted the PC gene was suspected. The translocation was validated via FISH-analysis and Sanger sequencing of its boundaries. Patient 2 is a 13 y.o. girl with psychomotor delay, episodes of lactic acidosis and ketonuria. WES revealed the novel homozygous intronic variant c.1983-116C > T. The PC's mRNA analysis demonstrated the exonization of several intron 16 sequences and some residual amount of WT mRNA isoform. Two other patients had more severe course of the disease. Their genotype represents missense variants in compound heterozygous and homozygous state (c.1876C > T (p.Arg626Trp), c.2606G > C (p.Gly869Ala), c.2435C > A (p.Ala812Asp). Conclusion In patients with metabolic crises, lactic acidosis and hypoglycemia analysis of PC gene is recommended. WGS with deep bioinformatic analysis should be taken into consideration when none or the only one pathogenic variant in the PC gene is found. In patients with metabolic crises with lactic acidosis and hypoglycemia WGS with deep bioinformatic analysis should be taken when none or the only one heterozygous pathogenic variant in the PC gene is found. PC gene may have more deep intronic pathogenic variants than included in known mutation databases.
Collapse
|
3
|
Assis AD, Chiarotto GB, da Silva NS, Simões GF, Oliveira ALR. Pregabalin synchronizes the regeneration of nerve and muscle fibers optimizing the gait recovery of MDX dystrophic mice. FASEB J 2022; 36:e22511. [PMID: 35998000 DOI: 10.1096/fj.202200411rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder induced by mutations in the dystrophin gene, leading to a degeneration of muscle fibers, triggering retrograde immunomodulatory, and degenerative events in the central nervous system. Thus, neuroprotective drugs such as pregabalin (PGB) can improve motor function by modulating plasticity, together with anti-inflammatory effects. The present work aimed to study the effects of PGB on axonal regeneration after axotomy in dystrophic and non-dystrophic mice. For that, MDX and C57BL/10 mouse strains were subjected to peripheral nerve damage and were treated with PGB (30 mg/kg/day, i.p.) for 28 consecutive days. The treatment was carried out in mice as soon as they completed 5 weeks of life, 1 week before the lesion, corresponding to the peak period of muscle degeneration in the MDX strain. Six-week-old mice were submitted to unilateral sciatic nerve crush and were sacrificed in the 9th week of age. The ipsi and contralateral sciatic nerves were processed for immunohistochemistry and qRT-PCR, evaluating the expression of proteins and gene transcripts related to neuronal and Schwann cell activity. Cranial tibial muscles were dissected for evaluation of neuromuscular junctions using α-bungarotoxin, and the myelinated axons of the sciatic nerve were analyzed by morphometry. The recovery of motor function was monitored throughout the treatment through tests of forced locomotion (rotarod) and spontaneous walking track test (Catwalk system). The results show that treatment with PGB reduced the retrograde cyclic effects of muscle degeneration/regeneration on the nervous system. This fact was confirmed after peripheral nerve injury, showing better adaptation and response of neurons and glia for rapid axonal regeneration, with efficient muscle targeting and regain of function. No side effects of PGB treatment were observed, and the expression of pro-regenerative proteins in neurons and Schwann cells was upregulated. Morphometry of the axons was in line with the preservation of motor endplates, resulting in enhanced performance of dystrophic animals. Overall, the present data indicate that pregabalin is protective and enhances regeneration of the SNP during the development of DMD, improving motor function, which can, in turn, be translated to the clinic.
Collapse
Affiliation(s)
- Alex Dias Assis
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Campinas, Brazil
| | | | | | | | | |
Collapse
|
4
|
Pregabalin-induced neuroprotection and gait improvement in dystrophic MDX mice. Mol Cell Neurosci 2021; 114:103632. [PMID: 34058345 DOI: 10.1016/j.mcn.2021.103632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease linked to the X chromosome induced by mutations in the dystrophin gene. Neuroprotective drugs, such as pregabalin (PGB), can improve motor function through the modulation of excitatory synapses, together with anti-apoptotic and anti-inflammatory effects. The present work studied the effects of PGB in the preservation of dystrophic peripheral nerves, allowing motor improvements in MDX mice. Five weeks old MDX and C57BL/10 mice were treated with PGB (30 mg/kg/day, i.p.) or vehicle, for 28 consecutive days. The mice were sacrificed on the 9th week, the sciatic nerves were dissected out and processed for immunohistochemistry and qRT-PCR, for evaluating the expression of proteins and gene transcripts related to neuronal activity and Schwann cell function. The lumbar spinal cords were also processed for qRT-PCR to evaluate the expression of neurotrophic factors and pro- and anti-inflammatory cytokines. Cranial tibial muscles were dissected out for endplate evaluation with α-bungarotoxin. The recovery of motor function was monitored throughout the treatment, using a spontaneous walking track test (Catwalk system) and a forced locomotion test (Rotarod). The results showed that treatment with PGB reduced the retrograde effects of muscle degeneration/regeneration on the nervous system from the 5th to the 9th week in MDX mice. Thus, PGB induced protein expression in neurons and Schwann cells, protecting myelinated fibers. In turn, better axonal morphology and close-to-normal motor endplates were observed. Indeed, such effects resulted in improved motor coordination of dystrophic animals. We believe that treatment with PGB improved the balance between excitatory and inhibitory inputs to spinal motoneurons, increasing motor control. In addition, PGB enhanced peripheral nerve homeostasis, by positively affecting Schwann cells. In general, the present results indicate that pregabalin is effective in protecting the PNS during the development of DMD, improving motor coordination, indicating possible translation to the clinic.
Collapse
|
5
|
Longo N, Dimmock D, Levy H, Viau K, Bausell H, Bilder DA, Burton B, Gross C, Northrup H, Rohr F, Sacharow S, Sanchez-Valle A, Stuy M, Thomas J, Vockley J, Zori R, Harding CO. Evidence- and consensus-based recommendations for the use of pegvaliase in adults with phenylketonuria. Genet Med 2018; 21:1851-1867. [PMID: 30546086 PMCID: PMC6752676 DOI: 10.1038/s41436-018-0403-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/29/2018] [Indexed: 11/23/2022] Open
Abstract
Purpose Phenylketonuria (PKU) is a rare metabolic disorder that requires
life-long management to reduce phenylalanine (Phe) concentrations within the
recommended range. The availability of pegvaliase (PALYNZIQ™, an enzyme that can
metabolize Phe) as a new therapy necessitates the provision of guidance for its
use. Methods A Steering Committee comprising 17 health-care professionals with
experience in using pegvaliase through the clinical development program drafted
guidance statements during a series of face-to-face meetings. A modified Delphi
methodology was used to demonstrate consensus among a wider group of health-care
professionals with experience in using pegvaliase. Results Guidance statements were developed for four categories: (1)
treatment goals and considerations prior to initiating therapy, (2) dosing
considerations, (3) considerations for dietary management, and (4) best
approaches to optimize medical management. A total of 34 guidance statements
were included in the modified Delphi voting and consensus was reached on all
after two rounds of voting. Conclusion Here we describe evidence- and consensus-based recommendations for
the use of pegvaliase in adults with PKU. The manuscript was evaluated against
the Appraisal of Guidelines for Research and Evaluation (AGREE II) instrument
and is intended for use by health-care professionals who will prescribe
pegvaliase and those who will treat patients receiving pegvaliase.
Collapse
Affiliation(s)
- Nicola Longo
- Division of Medical Genetics, University of Utah, Salt Lake City, UT, USA.
| | - David Dimmock
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
| | - Harvey Levy
- Division of Genetics and Genomics, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Krista Viau
- Division of Genetics and Genomics, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Heather Bausell
- Division of Clinical Nutrition & Genetics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Deborah A Bilder
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Barbara Burton
- Department of Medical Genetics, Ann & Robert H. Lurie Children's Hospital of Chicago and Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Christel Gross
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Hope Northrup
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fran Rohr
- Nutrition Center, Boston Children's Hospital, Boston, MA, USA
| | - Stephanie Sacharow
- Division of Genetics and Genomics, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | | | - Mary Stuy
- Department of Medical and Molecular Genetics, IU School of Medicine, Indianapolis, IN, USA
| | - Janet Thomas
- Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jerry Vockley
- Department of Pediatrics University of Pittsburgh School of Medicine, Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Roberto Zori
- Division of Genetics and Metabolism, University of Florida, Gainesville, FL, USA
| | - Cary O Harding
- Departments of Molecular and Medical Genetics and Pediatrics, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
6
|
Addicks GC, Marshall P, Jasmin BJ, Renaud JM, Zhang H, Menzies KJ. Critical Assessment of the mdx Mouse with Ex Vivo Eccentric Contraction of the Diaphragm Muscle. ACTA ACUST UNITED AC 2018; 8:e49. [PMID: 30106518 DOI: 10.1002/cpmo.49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Muscle function and health progressively deteriorate during the progression of muscle dystrophies. The ability to objectively characterize muscle function and muscle damage is useful not only when comparing variants of dystrophy models, but also for characterizing the effects of interventions aiming to improve or halt the progressive decline of muscle function and muscle health. The protocols in this chapter describe the use of ex vivo eccentric contraction of the diaphragm muscle as a measure of muscle susceptibility to damage. Because muscle has a robust regenerative capacity, unhealthy muscle may be functionally close to normal; therefore, protocols for ex vivo characterization of muscle are often essential for assessing the effects of interventions. Additional methods that can be applied for assessment of dystrophic muscle are also highlighted. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Gregory C Addicks
- Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Phillip Marshall
- Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Bernard J Jasmin
- Cellular and Molecular Medicine Department, Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-Marc Renaud
- Cellular and Molecular Medicine Department, Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, and The Department of Histology and Embryology of Zhongshan School of Medicine, Sun-Yat Sen University, Guangzhou, China
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Petrillo S, Pelosi L, Piemonte F, Travaglini L, Forcina L, Catteruccia M, Petrini S, Verardo M, D'Amico A, Musarò A, Bertini E. Oxidative stress in Duchenne muscular dystrophy: focus on the NRF2 redox pathway. Hum Mol Genet 2018; 26:2781-2790. [PMID: 28472288 DOI: 10.1093/hmg/ddx173] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/26/2017] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is involved in the pathogenesis of Duchenne muscular dystrophy (DMD), an X-linked genetic disorder caused by mutations in the dystrophin gene and characterized by progressive, lethal muscle degeneration and chronic inflammation. In this study, we explored the expression and signaling pathway of a master player of the anti-oxidant and anti-inflammatory response, namely NF-E2-related Factor 2, in muscle biopsies of DMD patients. We classified DMD patients in two age groups (Class I, 0-2 years and Class II, 2-9 years), in order to evaluate the antioxidant pathway expression during the disease progression. We observed that altered enzymatic antioxidant responses, increased levels of oxidized glutathione and oxidative damage are differently modulated in the two age classes of patients and well correlate with the severity of pathology. Interestingly, we also observed a modulation of relevant markers of the inflammatory response, such as heme oxygenase 1 and Inteleukin-6 (IL-6), suggesting a link between oxidative stress and chronic inflammatory response. Of note, using a transgenic mouse model, we demonstrated that IL-6 overexpression parallels the antioxidant expression profile and the severity of dystrophic muscle observed in DMD patients. This study advances our understanding of the pathogenic mechanisms underlying DMD and defines the critical role of oxidative stress on muscle wasting with clear implications for disease pathogenesis and therapy in human.
Collapse
Affiliation(s)
- Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| | - Laura Pelosi
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| | - Lorena Travaglini
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| | - Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Michela Catteruccia
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| | - Stefania Petrini
- Laboratory of Research, Children's Hospital and Research Institute Bambino Gesù, 00146 Rome, Italy
| | - Margherita Verardo
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| | - Adele D'Amico
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Enrico Bertini
- Unit of Muscular and Neurodegenerative Diseases, Children's Hospital and Research Institute Bambino Gesú, 00146 Rome, Italy
| |
Collapse
|
8
|
Luukkonen TM, Mehrjouy MM, Pöyhönen M, Anttonen A, Lahermo P, Ellonen P, Paulin L, Tommerup N, Palotie A, Varilo T. Breakpoint mapping and haplotype analysis of translocation t(1;12)(q43;q21.1) in two apparently independent families with vascular phenotypes. Mol Genet Genomic Med 2018; 6:56-68. [PMID: 29168350 PMCID: PMC5823676 DOI: 10.1002/mgg3.346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The risk of serious congenital anomaly for de novo balanced translocations is estimated to be at least 6%. We identified two apparently independent families with a balanced t(1;12)(q43;q21.1) as an outcome of a "Systematic Survey of Balanced Chromosomal Rearrangements in Finns." In the first family, carriers (n = 6) manifest with learning problems in childhood, and later with unexplained neurological symptoms (chronic headache, balance problems, tremor, fatigue) and cerebral infarctions in their 50s. In the second family, two carriers suffer from tetralogy of Fallot, one from transient ischemic attack and one from migraine. The translocation cosegregates with these vascular phenotypes and neurological symptoms. METHODS AND RESULTS We narrowed down the breakpoint regions using mate pair sequencing. We observed conserved haplotypes around the breakpoints, pointing out that this translocation has arisen only once. The chromosome 1 breakpoint truncates a CHRM3 processed transcript, and is flanked by the 5' end of CHRM3 and the 3' end of RYR2. TRHDE, KCNC2, and ATXN7L3B flank the chromosome 12 breakpoint. CONCLUSIONS This study demonstrates a balanced t(1;12)(q43;q21.1) with conserved haplotypes on the derived chromosomes. The translocation seems to result in vascular phenotype, with or without neurological symptoms, in at least two families. We suggest that the translocation influences the positional expression of CHRM3, RYR2, TRHDE, KCNC2, and/or ATXN7L3B.
Collapse
Affiliation(s)
- Tiia Maria Luukkonen
- Institute for molecular medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
- Department of HealthNational Institute for Health and WelfareHelsinkiFinland
| | - Mana M. Mehrjouy
- Wilhelm Johannsen Centre for Functional Genome ResearchDepartment of Cellular and Molecular MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Minna Pöyhönen
- Clinical GeneticsHelsinki University HospitalUniversity of HelsinkiHelsinkiFinland
- Department of Medical GeneticsUniversity of HelsinkiHelsinkiFinland
| | | | - Päivi Lahermo
- Institute for molecular medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Pekka Ellonen
- Institute for molecular medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
| | - Lars Paulin
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | - Niels Tommerup
- Wilhelm Johannsen Centre for Functional Genome ResearchDepartment of Cellular and Molecular MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Aarno Palotie
- Institute for molecular medicine Finland FIMMUniversity of HelsinkiHelsinkiFinland
- Broad Institute of Harvard and MITCambridgeMAUSA
| | - Teppo Varilo
- Department of HealthNational Institute for Health and WelfareHelsinkiFinland
- Department of Medical GeneticsUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
9
|
Wang L, Chen M, He R, Sun Y, Yang J, Xiao L, Cao J, Zhang H, Zhang C. Serum Creatinine Distinguishes Duchenne Muscular Dystrophy from Becker Muscular Dystrophy in Patients Aged ≤3 Years: A Retrospective Study. Front Neurol 2017; 8:196. [PMID: 28533764 PMCID: PMC5421192 DOI: 10.3389/fneur.2017.00196] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/21/2017] [Indexed: 11/13/2022] Open
Abstract
Here, we investigated correlations between serum creatinine (SCRN) levels and clinical phenotypes of dystrophinopathy in young patients. Sixty-eight patients with dystrophinopathy at the Neuromuscular Clinic, The First Affiliated Hospital, Sun Yat-sen University, were selected for this study. The diagnosis of dystrophinopathy was based on clinical manifestation, biochemical changes, and molecular analysis. Some patients underwent muscle biopsies; SCRN levels were tested when patients were ≤3 years old, and reading frame changes were analyzed. Each patient was followed up, and motor function and clinical phenotype were assessed when the same patients were ≥4 years old. Our findings indicated that in young patients, lower SCRN levels were associated with increased disease severity (p < 0.01) and that SCRN levels were the highest in patients exhibiting mild Becker muscular dystrophy (BMD) (p < 0.001) and the lowest in patients with Duchenne muscular dystrophy (DMD) (p < 0.01) and were significantly higher in patients carrying in-frame mutations than in patients carrying out-of-frame mutations (p < 0.001). SCRN level cutoff values for identifying mild BMD [18 µmol/L; area under the curve (AUC): 0.947; p < 0.001] and DMD (17 µmol/L; AUC: 0.837; p < 0.001) were established. These results suggest that SCRN might be a valuable biomarker for distinguishing DMD from BMD in patients aged ≤3 years and could assist in the selection of appropriate treatment strategies.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Menglong Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ruojie He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiming Sun
- Department of Health Care, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Juan Yang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lulu Xiao
- The Department of Tissue Typing Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiqing Cao
- Department of Neurology, Wuhan Central Hospital, Wuhan, Hubei, China
| | - Huili Zhang
- Department of Neurology, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Annese T, Corsi P, Ruggieri S, Tamma R, Marinaccio C, Picocci S, Errede M, Specchia G, De Luca A, Frassanito MA, Desantis V, Vacca A, Ribatti D, Nico B. Isolation and characterization of neural stem cells from dystrophic mdx mouse. Exp Cell Res 2016; 343:190-207. [DOI: 10.1016/j.yexcr.2016.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 10/22/2022]
|
11
|
Boussaïd G, Lofaso F, Santos DB, Vaugier I, Pottier S, Prigent H, Bahrami S, Orlikowski D. Impact of invasive ventilation on survival when non-invasive ventilation is ineffective in patients with Duchenne muscular dystrophy: A prospective cohort. Respir Med 2016; 115:26-32. [PMID: 27215500 DOI: 10.1016/j.rmed.2016.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Many patients with DMD undergo tracheostomy. Tracheostomy is associated with certain complications, however its effect on prognosis is not known. METHODS The relationship between type of mechanical ventilation and survival at 12 years was evaluated in a prospective cohort of patients with Duchenne muscular dystrophy followed in a French reference center for Neuromuscular Diseases. Cox proportional-hazards regressions were used to estimate the hazard ratios associated with risk of switching from non-invasive to invasive ventilation, and with risk of death. RESULTS One hundred and fifty patients were included. Initial use of invasive ventilation was associated with an episode of acute respiratory failure (p < 0.0001) and with a severe clinical status (p < 0.05). Risk of death was associated with swallowing disorders (2.51, IC [1.12-5.66], p < 0.03) and cardiac failure (p < 0.05) but not with type of mechanical ventilation. CONCLUSION Switching to invasive ventilation is appropriate when non-invasive ventilation is ineffective.
Collapse
Affiliation(s)
- Ghilas Boussaïd
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France.
| | - Frédéric Lofaso
- Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Service d'Explorations Fonctionnelles Respiratoires, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - Dante Brasil Santos
- Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Service d'Explorations Fonctionnelles Respiratoires, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - Isabelle Vaugier
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - Sandra Pottier
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - Hélène Prigent
- Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Service d'Explorations Fonctionnelles Respiratoires, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - Stéphane Bahrami
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Université de Versailles Saint Quentin en Yvelines, EA 4047, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Service de Santé Publique, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| | - David Orlikowski
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France; Service de Santé Publique, AP-HP, Hôpital Raymond Poincaré, 92380, Garches, France
| |
Collapse
|
12
|
Lala-Tabbert N, Fu D, Wiper-Bergeron N. Induction of CCAAT/Enhancer-Binding Protein β Expression With the Phosphodiesterase Inhibitor Isobutylmethylxanthine Improves Myoblast Engraftment Into Dystrophic Muscle. Stem Cells Transl Med 2016; 5:500-10. [PMID: 26941360 DOI: 10.5966/sctm.2015-0169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/25/2015] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is the most common muscular dystrophy. Characterized by rounds of muscle degeneration and regeneration, DMD features progressive muscle wasting and is fatal. One approach for treatment is transplantation of muscle progenitor cells to repair and restore dystrophin expression to damaged muscle. However, the success of this approach has been limited by difficulties in isolating large numbers of myogenic progenitors with strong regenerative potential, poor engraftment, poor survival of donor cells, and limited migration in the diseased muscle. We demonstrate that induction of the transcription factor CCAAT/enhancer-binding protein β (C/EBPβ) using the cyclic adenosine monophosphate phosphodiesterase inhibitor isobutylmethylxanthine (IBMX) results in enhanced myoblast expansion in culture and increased satellite cell marker expression. When equal numbers of IBMX-treated cells were transplanted into dystrophic muscle, they contributed to muscle repair more efficiently than did vehicle-treated cells and engrafted into the satellite cell niche in higher numbers, demonstrating improved cell migration from the site of injury and enhanced survival after transplantation. Thus, pharmacologic stimulation of C/EBPβ expression reprograms myoblasts to a more stem cell-like state, promotes expansion in culture, and improves engraftment such that better transplantation outcomes are achieved. SIGNIFICANCE Duchenne muscular dystrophy is a genetic disorder for which no cure exists. One therapeutic approach is transplantation of myogenic progenitors to restore dystrophin to damaged muscle, but this approach is limited by poor engraftment of cultured myoblasts. Transient upregulation of CCAAT/enhancer-binding protein β in primary myoblasts using the phosphodiesterase isobutylmethylxanthine (IBMX) increases satellite cell marker expression in cultured myoblasts, improves their migration, and increases their survival after transplantation. When transplanted into C57BL/10ScSn-mdx/J mice , IBMX-treated myoblasts restored dystrophin expression and were able to occupy the satellite cell niche more efficiently than controls. A myoblast culture approach that reprograms myoblasts to a more primitive state, resulting in improved transplantation outcomes and reinvigorating research into myoblast transplantation as a viable therapeutic approach, is described.
Collapse
Affiliation(s)
- Neena Lala-Tabbert
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dechen Fu
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
13
|
2015 William Allan Award. Am J Hum Genet 2016; 98:419-426. [PMID: 26942278 DOI: 10.1016/j.ajhg.2016.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Indexed: 11/21/2022] Open
|
14
|
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy are caused by mutations in the dystrophin-encoding DMD gene. Large deletions and duplications are most common, but small mutations have been found as well. Having a correct diagnosis is important for family planning and providing proper care to patients according to published guidelines. With mutation-specific therapies under development for DMD, a correct diagnosis is now also important for assessing whether patients are eligible for treatments. This review discusses different mutations causing DMD, diagnostic techniques available for making a genetic diagnosis for children suspected of DMD and the importance of having a specific genetic diagnosis in the context of emerging genetic therapies for DMD.
Collapse
Affiliation(s)
- Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands,John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Ieke B Ginjaar
- Laboratory for Diagnostics Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| |
Collapse
|
15
|
Aartsma-Rus A, Ginjaar IB, Bushby K. The importance of genetic diagnosis for Duchenne muscular dystrophy. J Med Genet 2016; 53:145-51. [PMID: 26754139 PMCID: PMC4789806 DOI: 10.1136/jmedgenet-2015-103387] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/10/2015] [Indexed: 12/18/2022]
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy are caused by mutations in the dystrophin-encoding DMD gene. Large deletions and duplications are most common, but small mutations have been found as well. Having a correct diagnosis is important for family planning and providing proper care to patients according to published guidelines. With mutation-specific therapies under development for DMD, a correct diagnosis is now also important for assessing whether patients are eligible for treatments. This review discusses different mutations causing DMD, diagnostic techniques available for making a genetic diagnosis for children suspected of DMD and the importance of having a specific genetic diagnosis in the context of emerging genetic therapies for DMD.
Collapse
Affiliation(s)
- Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Ieke B Ginjaar
- Laboratory for Diagnostics Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| |
Collapse
|
16
|
Guiraud S, Aartsma-Rus A, Vieira NM, Davies KE, van Ommen GJB, Kunkel LM. The Pathogenesis and Therapy of Muscular Dystrophies. Annu Rev Genomics Hum Genet 2015; 16:281-308. [DOI: 10.1146/annurev-genom-090314-025003] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Simon Guiraud
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, OX1 3PT Oxford, United Kingdom; ,
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; ,
| | - Natassia M. Vieira
- Division of Genetics and Genomics and Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Kay E. Davies
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, OX1 3PT Oxford, United Kingdom; ,
| | - Gert-Jan B. van Ommen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; ,
| | - Louis M. Kunkel
- Division of Genetics and Genomics and Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts 02115
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| |
Collapse
|
17
|
Aristizabal JF, Smit RM. Orthodontic treatment in a patient with unilateral open-bite and Becker muscular dystrophy. A 5-year follow-up. Dental Press J Orthod 2015; 19:37-45. [PMID: 25628078 PMCID: PMC4347409 DOI: 10.1590/2176-9451.19.6.037-045.oar] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 11/01/2013] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION: Becker muscular dystrophy is an X-chromosomal linked anomaly characterized by
progressive muscle wear and weakness. This case report shows the orthodontic
treatment of a Becker muscular dystrophy patient with unilateral open bite. METHODS: To correct patient's malocclusion, general anesthesia and orthognathic surgery
were not considered as an option. Conventional orthodontic treatment with
intermaxillary elastics and muscular functional therapy were employed instead.
RESULTS: After 36 months, open bite was corrected. The case remains stable after a 5-year
post-treatment retention period.
Collapse
|
18
|
Goudot FX, Wahbi K, Aïssou L, Sorbets E, Siam-Tsieu V, Eymard B, Themar Noel C, Devaux JY, Dessault O, Duboc D, Meune C. Reduced inotropic reserve is predictive of further degradation in left ventricular ejection fraction in patients with Duchenne muscular dystrophy. Eur J Heart Fail 2014; 17:177-81. [DOI: 10.1002/ejhf.213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
| | - Karim Wahbi
- Paris Descartes University; APHP, Department of Cardiology, Cochin Hospital; Paris France
- Myology Institute; APHP, Pitié-Salpêtrière Hospital; Paris France
| | - Linda Aïssou
- Paris 13 University, Avicenne Hospital; Department of Cardiology; APHP Bobigny France
| | - Emmanuel Sorbets
- Paris 13 University, Avicenne Hospital; Department of Cardiology; APHP Bobigny France
- Department of Cardiology; Bichat Hospital; APHP Paris France
| | - Valérie Siam-Tsieu
- Paris Descartes University; APHP, Department of Cardiology, Cochin Hospital; Paris France
| | - Bruno Eymard
- Myology Institute; APHP, Pitié-Salpêtrière Hospital; Paris France
| | | | - Jean-Yves Devaux
- Saint-Antoine Hospital; Department of Nuclear Medicine; Paris France
| | - Odile Dessault
- Saint-Antoine Hospital; Department of Nuclear Medicine; Paris France
| | - Denis Duboc
- Paris Descartes University; APHP, Department of Cardiology, Cochin Hospital; Paris France
| | - Christophe Meune
- Paris 13 University, Avicenne Hospital; Department of Cardiology; APHP Bobigny France
- Paris Descartes University; APHP, Department of Cardiology, Cochin Hospital; Paris France
- INSERM UMR S-942; Paris France
| |
Collapse
|
19
|
Pillers DAM. A new day for Duchenne's?: The time has come for newborn screening. Mol Genet Metab 2014; 113:11-3. [PMID: 24974086 DOI: 10.1016/j.ymgme.2014.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 06/07/2014] [Indexed: 01/16/2023]
Affiliation(s)
- De-Ann M Pillers
- Department of Pediatrics, University of Wisconsin, Madison, USA.
| |
Collapse
|
20
|
Utami KH, Hillmer AM, Aksoy I, Chew EGY, Teo ASM, Zhang Z, Lee CWH, Chen PJ, Seng CC, Ariyaratne PN, Rouam SL, Soo LS, Yousoof S, Prokudin I, Peters G, Collins F, Wilson M, Kakakios A, Haddad G, Menuet A, Perche O, Tay SKH, Sung KWK, Ruan X, Ruan Y, Liu ET, Briault S, Jamieson RV, Davila S, Cacheux V. Detection of chromosomal breakpoints in patients with developmental delay and speech disorders. PLoS One 2014; 9:e90852. [PMID: 24603971 PMCID: PMC3946304 DOI: 10.1371/journal.pone.0090852] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 02/04/2014] [Indexed: 01/25/2023] Open
Abstract
Delineating candidate genes at the chromosomal breakpoint regions in the apparently balanced chromosome rearrangements (ABCR) has been shown to be more effective with the emergence of next-generation sequencing (NGS) technologies. We employed a large-insert (7-11 kb) paired-end tag sequencing technology (DNA-PET) to systematically analyze genome of four patients harbouring cytogenetically defined ABCR with neurodevelopmental symptoms, including developmental delay (DD) and speech disorders. We characterized structural variants (SVs) specific to each individual, including those matching the chromosomal breakpoints. Refinement of these regions by Sanger sequencing resulted in the identification of five disrupted genes in three individuals: guanine nucleotide binding protein, q polypeptide (GNAQ), RNA-binding protein, fox-1 homolog (RBFOX3), unc-5 homolog D (C.elegans) (UNC5D), transmembrane protein 47 (TMEM47), and X-linked inhibitor of apoptosis (XIAP). Among them, XIAP is the causative gene for the immunodeficiency phenotype seen in the patient. The remaining genes displayed specific expression in the fetal brain and have known biologically relevant functions in brain development, suggesting putative candidate genes for neurodevelopmental phenotypes. This study demonstrates the application of NGS technologies in mapping individual gene disruptions in ABCR as a resource for deciphering candidate genes in human neurodevelopmental disorders (NDDs).
Collapse
Affiliation(s)
- Kagistia H. Utami
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Axel M. Hillmer
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, Singapore
| | - Irene Aksoy
- Stem Cells and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Elaine G. Y. Chew
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, Singapore
| | - Audrey S. M. Teo
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, Singapore
| | - Zhenshui Zhang
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, Singapore
| | - Charlie W. H. Lee
- Computational and Mathematical Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Pauline J. Chen
- Computational and Mathematical Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Chan Chee Seng
- Scientific & Research Computing, Genome Institute of Singapore, Singapore, Singapore
| | - Pramila N. Ariyaratne
- Computational and Mathematical Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Sigrid L. Rouam
- Computational and Mathematical Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Lim Seong Soo
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Saira Yousoof
- Eye and Developmental Genetics Research, The Children’s Hospital at Westmead, Children’s Medical Research Institute and Save Sight Institute, Sydney, New South Wales, Australia
- Disciplines of Paediatrics and Child Health and Genetic Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Ivan Prokudin
- Eye and Developmental Genetics Research, The Children’s Hospital at Westmead, Children’s Medical Research Institute and Save Sight Institute, Sydney, New South Wales, Australia
- Disciplines of Paediatrics and Child Health and Genetic Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Gregory Peters
- Department of Cytogenetics, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Felicity Collins
- Department of Clinical Genetics, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Meredith Wilson
- Department of Clinical Genetics, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Alyson Kakakios
- Department of Immunology, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | | | - Arnaud Menuet
- Service de Genetique INEM UMR7355 CNRS-University, Centre Hospitalier Régional d’Orléans, Orléans, France
| | - Olivier Perche
- Service de Genetique INEM UMR7355 CNRS-University, Centre Hospitalier Régional d’Orléans, Orléans, France
| | - Stacey Kiat Hong Tay
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ken W. K. Sung
- Computational and Mathematical Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Xiaoan Ruan
- Genome Technology and Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Yijun Ruan
- Genome Technology and Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Edison T. Liu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, Singapore
| | - Sylvain Briault
- Service de Genetique INEM UMR7355 CNRS-University, Centre Hospitalier Régional d’Orléans, Orléans, France
| | - Robyn V. Jamieson
- Eye and Developmental Genetics Research, The Children’s Hospital at Westmead, Children’s Medical Research Institute and Save Sight Institute, Sydney, New South Wales, Australia
| | - Sonia Davila
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Valere Cacheux
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
21
|
Barnabei MS, Martindale JM, Townsend D, Metzger JM. Exercise and muscular dystrophy: implications and analysis of effects on musculoskeletal and cardiovascular systems. Compr Physiol 2013; 1:1353-63. [PMID: 23733645 DOI: 10.1002/cphy.c100062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The muscular dystrophies are a heterogeneous collection of progressive, inherited diseases of muscle weakness and degeneration. Although these diseases can vary widely in their etiology and presentation, nearly all muscular dystrophies cause exercise intolerance to some degree. Here, we focus on Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, as a paradigm for the effects of muscle disease on exercise capacity. First described in the mid-1800s, DMD is a rapidly progressive and lethal muscular dystrophy caused by mutations in the dystrophin gene. Dystrophin is a membrane-associated cytoskeletal protein, the loss of which causes numerous cellular defects including mechanical instability of the sarcolemma, increased influx of extracellular calcium, and cell signaling defects. Here, we discuss the physiological basis for exercise intolerance in DMD, focusing on the molecular and cellular defects caused by loss of dystrophin and how these manifest as organ-level dysfunction and reduced exercise capacity. The main focus of this article is the defects present in dystrophin-deficient striated muscle. However, discussion regarding the effects of dystrophin loss on other tissues, including vascular smooth muscle is also included. Collectively, the goal of this article is to summarize the current state of knowledge regarding the mechanistic basis for exercise intolerance in DMD, which may serve as an archetype for other muscular dystrophies and diseases of muscle wasting.
Collapse
Affiliation(s)
- Matthew S Barnabei
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | | | | | | |
Collapse
|
22
|
Cripe LH, Tobias JD. Cardiac considerations in the operative management of the patient with Duchenne or Becker muscular dystrophy. Paediatr Anaesth 2013; 23:777-84. [PMID: 23869433 DOI: 10.1111/pan.12229] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2013] [Indexed: 12/22/2022]
Abstract
Duchenne muscular dystrophy/Becker muscular dystrophy (DMD/BMD) is a progressive multisystem neuromuscular disorder. In addition to the skeletal muscle, the myocardium in the DMD/BMD patient is dystrophin deficient which results in a progressive cardiomyopathy. The myopathic myocardium poses significant risk of increased morbidity and mortality at the time of major surgical procedures. Careful attention must be given to the DMD/BMD patient during the intraoperative and postoperative period. Anesthesia selection is critical and anesthetics should be avoided which have been shown to be harmful in this patient population. Preanesthesia assessment should include cardiac consultation and detailed preoperative evaluation. Intraoperative management needs to insure that the weakened myocardium is not compromised by physiologic changes such as hypotension or major fluid shifts. Finally, attention to the cardiac status of the patient must continue into the postoperative period. The surgical care of the DMD/BMD patient requires a multispecialty approach to insure operative success.
Collapse
Affiliation(s)
- Linda H Cripe
- The Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH 43205-2664, USA.
| | | |
Collapse
|
23
|
Shigemoto T, Kuroda Y, Wakao S, Dezawa M. A novel approach to collecting satellite cells from adult skeletal muscles on the basis of their stress tolerance. Stem Cells Transl Med 2013; 2:488-98. [PMID: 23748608 DOI: 10.5966/sctm.2012-0130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Stem cells are generally collected using flow cytometry, but this method is not applicable when the cell surface marker is not well determined. Satellite cells, which are skeletal muscle stem cells, have the ability to regenerate damaged muscles and are expected to be applicable for treatment of muscle degeneration. Although the transcription factor Pax7 is a known specific marker of satellite cells, it is not located on the cell surface and therefore flow cytometry is not directly applicable. In the present study, we turned our attention to the stress tolerance of adult stem cells, and we propose long-term trypsin incubation (LTT) as a novel approach to collecting satellite cells from mouse and human skeletal muscles. LTT led to a remarkable increase in the ratio of Pax7(+) cells that retain normal myogenic stem cell function. In particular, human Pax7(+) cells made up approximately 30% of primary cultured cells, whereas after LTT, the ratio of Pax7(+) cells increased up to ∼80%, and the ratio of Pax7(+) and/or MyoD(+) myogenic cells increased to ∼95%. Once transplanted, LTT-treated cells contributed to subsequent muscle regeneration following repetitive muscle damage without additional cell transplantation. The stress tolerance of Pax7(+) cells is related to heat shock protein 27 and αB-crystallin, members of the small heat shock protein family. This approach, based on the stress resistance of adult stem cells, is a safe and inexpensive method of efficiently collecting human satellite cells and may also be used for collecting other tissue stem cells whose surface marker is unknown.
Collapse
Affiliation(s)
- Taeko Shigemoto
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| | | | | | | |
Collapse
|
24
|
Meregalli M, Farini A, Belicchi M, Parolini D, Cassinelli L, Razini P, Sitzia C, Torrente Y. Perspectives of stem cell therapy in Duchenne muscular dystrophy. FEBS J 2013. [DOI: 10.1111/febs.12083] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Mirella Meregalli
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Andrea Farini
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Marzia Belicchi
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Daniele Parolini
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Letizia Cassinelli
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Paola Razini
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Clementina Sitzia
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| | - Yvan Torrente
- Laboratorio Cellule Staminali; Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti; Università degli Studi di Milano; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Milan; Italy
| |
Collapse
|
25
|
Ernste FC, Reed AM. Idiopathic inflammatory myopathies: current trends in pathogenesis, clinical features, and up-to-date treatment recommendations. Mayo Clin Proc 2013; 88:83-105. [PMID: 23274022 DOI: 10.1016/j.mayocp.2012.10.017] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 10/30/2012] [Accepted: 10/31/2012] [Indexed: 01/13/2023]
Abstract
Recently, there have been important advances in the understanding of the pathophysiologic features, assessment, and management of patients with a newly diagnosed idiopathic inflammatory myopathy (IIM). Myositis-specific autoantibodies have been identified to define patient subgroups and offer prognostic implications. Similarly, proinflammatory cytokines, such as interleukin 6 and type 1 interferon-dependent genes, may serve as potential biomarkers of disease activity in adult and juvenile patients with dermatomyositis (DM). Moreover, magnetic resonance imaging has become an important modality for the assessment of muscle inflammation in adult IIM and juvenile DM. Immune-mediated necrotizing myopathies also are being recognized as a subset of IIM triggered by medications such as statins. However, confusion exists regarding effective management strategies for patients with IIM because of the lack of large-scale, randomized, controlled studies. This review focuses primarily on our current management and treatment algorithms for IIM including the care of pediatric patients with juvenile DM. For this review, we conducted a search of PubMed and MEDLINE for articles published from January 1, 1970, to December 1, 2011, using the following search terms: idiopathic inflammatory myopathies, dermatomyositis, polymyositis, juvenile dermatomyositis, sporadic inclusion body myositis, inclusion body myositis, inflammatory myositis, myositis, myopathies, pathogenesis, therapy, and treatment. Studies published in English were selected for inclusion in our review as well as additional articles identified from bibliographies.
Collapse
|
26
|
Seto JT, Ramos JN, Muir L, Chamberlain JS, Odom GL. Gene replacement therapies for duchenne muscular dystrophy using adeno-associated viral vectors. Curr Gene Ther 2012; 12:139-51. [PMID: 22533379 DOI: 10.2174/156652312800840603] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 04/16/2012] [Accepted: 04/17/2012] [Indexed: 12/12/2022]
Abstract
The muscular dystrophies collectively represent a major health challenge, as few significant treatment options currently exist for any of these disorders. Recent years have witnessed a proliferation of novel approaches to therapy, spanning increased testing of existing and new pharmaceuticals, DNA delivery (both anti-sense oligonucleotides and plasmid DNA), gene therapies and stem cell technologies. While none of these has reached the point of being used in clinical practice, all show promise for being able to impact different types of muscular dystrophies. Our group has focused on developing direct gene replacement strategies to treat recessively inherited forms of muscular dystrophy, particularly Duchenne and Becker muscular dystrophy (DMD/BMD). Both forms of dystrophy are caused by mutations in the dystrophin gene and all cases can in theory be treated by gene replacement using synthetic forms of the dystrophin gene. The major challenges for success of this approach are the development of a suitable gene delivery shuttle, generating a suitable gene expression cassette able to be carried by such a shuttle, and achieving safe and effective delivery without elicitation of a destructive immune response. This review summarizes the current state of the art in terms of using adeno-associated viral vectors to deliver synthetic dystrophin genes for the purpose of developing gene therapy for DMD.
Collapse
Affiliation(s)
- Jane T Seto
- Department of Neurology, University of Washington, Seattle, WA 98195-7720, USA.
| | | | | | | | | |
Collapse
|
27
|
Weisleder N, Takizawa N, Lin P, Wang X, Cao C, Zhang Y, Tan T, Ferrante C, Zhu H, Chen PJ, Yan R, Sterling M, Zhao X, Hwang M, Takeshima M, Cai C, Cheng H, Takeshima H, Xiao RP, Ma J. Recombinant MG53 protein modulates therapeutic cell membrane repair in treatment of muscular dystrophy. Sci Transl Med 2012; 4:139ra85. [PMID: 22723464 DOI: 10.1126/scitranslmed.3003921] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Mitsugumin 53 (MG53), a muscle-specific TRIM family protein, is an essential component of the cell membrane repair machinery. Here, we examined the translational value of targeting MG53 function in tissue repair and regenerative medicine. Although native MG53 protein is principally restricted to skeletal and cardiac muscle tissues, beneficial effects that protect against cellular injuries are present in nonmuscle cells with overexpression of MG53. In addition to the intracellular action of MG53, injury to the cell membrane exposes a signal that can be detected by MG53, allowing recombinant MG53 protein to repair membrane damage when provided in the extracellular space. Recombinant human MG53 (rhMG53) protein purified from Escherichia coli fermentation provided dose-dependent protection against chemical, mechanical, or ultraviolet-induced damage to both muscle and nonmuscle cells. Injection of rhMG53 through multiple routes decreased muscle pathology in the mdx dystrophic mouse model. Our data support the concept of targeted cell membrane repair in regenerative medicine, and present MG53 protein as an attractive biological reagent for restoration of membrane repair defects in human diseases.
Collapse
Affiliation(s)
- Noah Weisleder
- Department of Physiology and Biophysics, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
OKA A, MABUCHI T, OZAWA A, INOKO H. Current understanding of human genetics and genetic analysis of psoriasis. J Dermatol 2012; 39:231-41. [DOI: 10.1111/j.1346-8138.2012.01504.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
29
|
Abstract
Structural variation, whether it is caused by copy number variants or present in a balanced form, such as reciprocal translocations and inversions, can have a profound and dramatic effect on the expression of genes mapping within and close to the rearrangement, as well as affecting others genome wide. These effects can be caused by altering the copy number of one or more genes or regulatory elements (dosage effect) or from physical disruption of links between regulatory elements and their associated gene or genes, resulting in perturbation of expression. Similarly, large-scale structural variants can result in genome-wide expression changes by altering the positions that chromosomes occupy within the nucleus, potentially disrupting not only local cis interactions, but also trans interactions that occur throughout the genome. Structural variation is, therefore, a significant factor in the study of gene expression and is discussed here in more detail.
Collapse
Affiliation(s)
- Louise Harewood
- The Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
30
|
Hor KN, Mazur W, Taylor MD, Al-Khalidi HR, Cripe LH, Jefferies JL, Raman SV, Chung ES, Kinnett KJ, Williams K, Gottliebson WM, Benson DW. Effects of steroids and angiotensin converting enzyme inhibition on circumferential strain in boys with Duchenne muscular dystrophy: a cross-sectional and longitudinal study utilizing cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2011; 13:60. [PMID: 22011358 PMCID: PMC3207955 DOI: 10.1186/1532-429x-13-60] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 10/19/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Steroid use has prolonged ambulation in Duchenne muscular dystrophy (DMD) and combined with advances in respiratory care overall management has improved such that cardiac manifestations have become the major cause of death. Unfortunately, there is no consensus for DMD-associated cardiac disease management. Our purpose was to assess effects of steroid use alone or in combination with angiotensin converting enzyme inhibitors (ACEI) or angiotension receptor blocker (ARB) on cardiovascular magnetic resonance (CMR) derived circumferential strain (εcc). METHODS We used CMR to assess effects of corticosteroids alone (Group A) or in combination with ACEI or ARB (Group B) on heart rate (HR), left ventricular ejection fraction (LVEF), mass (LVM), end diastolic volume (LVEDV) and circumferential strain (εcc) in a cohort of 171 DMD patients >5 years of age. Treatment decisions were made independently by physicians at both our institution and referral centers and not based on CMR results. RESULTS Patients in Group A (114 studies) were younger than those in Group B (92 studies)(10 ± 2.4 vs. 12.4 ± 3.2 years, p < 0.0001), but HR, LVEF, LVEDV and LVM were not different. Although εcc magnitude was lower in Group B than Group A (-13.8 ± 1.9 vs. -12.8 ± 2.0, p = 0.0004), age correction using covariance analysis eliminated this effect. In a subset of patients who underwent serial CMR exams with an inter-study time of ~15 months, εcc worsened regardless of treatment group. CONCLUSIONS These results support the need for prospective clinical trials to identify more effective treatment regimens for DMD associated cardiac disease.
Collapse
Affiliation(s)
- Kan N Hor
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Wojciech Mazur
- The Heart and Vascular Center at The Christ Hospitals, Cincinnati, Ohio, USA
| | - Michael D Taylor
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Linda H Cripe
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - John L Jefferies
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Eugene S Chung
- The Heart and Vascular Center at The Christ Hospitals, Cincinnati, Ohio, USA
| | - Kathi J Kinnett
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katelyn Williams
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - William M Gottliebson
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - D Woodrow Benson
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
31
|
James J, Kinnett K, Wang Y, Ittenbach RF, Benson DW, Cripe L. Electrocardiographic abnormalities in very young Duchenne muscular dystrophy patients precede the onset of cardiac dysfunction. Neuromuscul Disord 2011; 21:462-7. [DOI: 10.1016/j.nmd.2011.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 03/14/2011] [Accepted: 04/15/2011] [Indexed: 10/18/2022]
|
32
|
Hor KN, Wansapura JP, Al-Khalidi HR, Gottliebson WM, Taylor MD, Czosek RJ, Nagueh SF, Akula N, Chung ES, Benson WD, Mazur W. Presence of mechanical dyssynchrony in Duchenne muscular dystrophy. J Cardiovasc Magn Reson 2011; 13:12. [PMID: 21288342 PMCID: PMC3041675 DOI: 10.1186/1532-429x-13-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 02/02/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac dysfunction in boys with Duchenne muscular dystrophy (DMD) is a leading cause of death. Cardiac resynchronization therapy (CRT) has been shown to dramatically decrease mortality in eligible adult population with congestive heart failure. We hypothesized that mechanical dyssynchrony is present in DMD patients and that cardiovascular magnetic resonance (CMR) may predict CRT efficacy. METHODS DMD patients (n = 236) were stratified into 4 groups based on age, diagnosis of DMD, left ventricular (LV) ejection fraction (EF), and presence of myocardial fibrosis defined as positive late gadolinum enhancement (LGE) compared to normal controls (n = 77). Dyssynchrony indices were calculated based on timing of CMR derived circumferential strain (ecc). The calculated indices included cross-correlation delay (XCD), uniformity of strain (US), regional vector of variance (RVV), time to maximum strain (TTMS) and standard deviation (SD) of TTMS. Abnormal XCD value was defined as > normal + 2SD. US, RVV, TTMS and SD were calculated for patients with abnormal XCD. RESULTS There was overall low prevalence of circumferential dyssynchrony in the entire DMD population; it increased to 17.1% for patients with abnormal EF and to 31.2% in the most advanced stage (abnormal EF with fibrosis). All but one DMD patient with mechanical dyssynchrony exhibited normal QRS duration suggesting absence of electrical dyssynchrony. The calculated US and RVV values (0.91 ± 0.09, 1.34 ± 0.48) indicate disperse rather than clustered dyssynchrony. CONCLUSION Mechanical dyssynchrony is frequent in boys with end stage DMD-associated cardiac dysfunction. It is associated with normal QRS complex as well as extensive lateral fibrosis. Based on these findings, it is unlikely that this patient population will benefit from CRT.
Collapse
Affiliation(s)
- Kan N Hor
- The Heart Institute and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Janaka P Wansapura
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - William M Gottliebson
- The Heart Institute and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Michael D Taylor
- The Heart Institute and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Richard J Czosek
- The Heart Institute and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Nandakishore Akula
- The Heart Institute and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Eugene S Chung
- The Heart and Vascular Center, The Christ Hospital, Cincinnati, Ohio, USA
| | - Woodrow D Benson
- The Heart Institute and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Wojciech Mazur
- The Heart and Vascular Center, The Christ Hospital, Cincinnati, Ohio, USA
| |
Collapse
|
33
|
Mazur W, Hor KN, Germann JT, Fleck RJ, Al-Khalidi HR, Wansapura JP, Chung ES, Taylor MD, Jefferies JL, Benson DW, Gottliebson WM. Patterns of left ventricular remodeling in patients with Duchenne Muscular Dystrophy: a cardiac MRI study of ventricular geometry, global function, and strain. Int J Cardiovasc Imaging 2011; 28:99-107. [PMID: 21222036 DOI: 10.1007/s10554-010-9781-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/22/2010] [Indexed: 01/02/2023]
Abstract
The cardiac disease ubiquitously associated in Duchenne Muscular Dystrophy (DMD) has traditionally been considered a progressive dilated cardiomyopathy (DCM). However, left ventricular (LV) dilatation as measured with cardiac MRI has not been a consistent finding in this population, even as circumferential strain (ε(cc)) declines with advancing disease. We hypothesized that a distinct pattern of changes in LV geometry, during the course of ε(cc) decline, distinguishes DMD associated heart disease from DCM. Using CMR, LV end-diastolic volume (EDV), mass (LVM), ejection fraction, ε(cc) and myocardial delayed enhancement (MDE) were determined in DMD patients and normal control subjects. The LV Remodeling Index (LVRI) was calculated as the ratio of LV Mass to Volume (LVM/EDV). Statistical comparisons between all LV parameters and genotype were also performed. Median LVRI in DMD (n = 127) and control subjects (n = 41) were different (0.75 vs. 0.65, P = 0.0150) but within normal range. Furthermore, the median LVRI in DMD boys with reduced LV systolic function was significantly reduced compared to those with normal LV systolic function (0.64 vs. 0.75, P = 0.0974). However, the presence of MDE was associated with a lower median LVRI (0.57 vs. 0.76, P = 0.0471). Regression analysis showed no significant correlation between ε(cc) and LVRI (r = -0.03). The LVRI of DMD patients is unexpectedly normal and not correlated with ε(cc.) Based on these findings, DMD-associated heart disease exhibits a unique remodeling pattern distinct from DCM.
Collapse
Affiliation(s)
- Wojciech Mazur
- The Heart and Vascular Center at the Christ Hospital, Cincinnati, OH, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Meregalli M, Farini A, Belicchi M, Torrente Y. CD133(+) cells isolated from various sources and their role in future clinical perspectives. Expert Opin Biol Ther 2010; 10:1521-8. [PMID: 20932225 DOI: 10.1517/14712598.2010.528386] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND CD133 is a member of a novel family of cell surface glycoproteins. Initially, the expression of CD133 antigen was seen only in the hematopoietic derived CD34(+) stem cells. At present, CD133 expression is demonstrated in undifferentiated epithelium, different types of tumors and myogenic cells. CD133(+) neurosphere cells isolated from brain are able to differentiate into both neurons and glial cells. These data suggested that CD133 could be a specific marker for various stem and progenitor cell populations. OBJECTIVES The main goal would be to describe the role for CD133 as a marker of stem cells able to engraft and differentiate, to form functional non-hematopoietic adult lineages and contribute to disease amelioration via tissue regeneration. RESULTS/CONCLUSION In conclusion, since the rise of CD133 antigen as a suitable stem cell marker, the possible use of CD133(+) stem cells in therapeutic applications has opened a new promising field in the treatment of degenerating diseases. The human circulating cells expressing the CD133 antigen behave as a stem cell population capable of commitment to hematopoietic, endothelial and myogenic lineages. CD133 cell therapy may represent a promising treatment for many diseases.
Collapse
Affiliation(s)
- Mirella Meregalli
- Stem Cell Laboratory, Department of Neurological Sciences, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Italy.
| | | | | | | |
Collapse
|
35
|
Mutations in GRIN2A and GRIN2B encoding regulatory subunits of NMDA receptors cause variable neurodevelopmental phenotypes. Nat Genet 2010; 42:1021-6. [PMID: 20890276 DOI: 10.1038/ng.677] [Citation(s) in RCA: 387] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 08/30/2010] [Indexed: 02/07/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors mediate excitatory neurotransmission in the mammalian brain. Two glycine-binding NR1 subunits and two glutamate-binding NR2 subunits each form highly Ca²(+)-permeable cation channels which are blocked by extracellular Mg²(+) in a voltage-dependent manner. Either GRIN2B or GRIN2A, encoding the NMDA receptor subunits NR2B and NR2A, was found to be disrupted by chromosome translocation breakpoints in individuals with mental retardation and/or epilepsy. Sequencing of GRIN2B in 468 individuals with mental retardation revealed four de novo mutations: a frameshift, a missense and two splice-site mutations. In another cohort of 127 individuals with idiopathic epilepsy and/or mental retardation, we discovered a GRIN2A nonsense mutation in a three-generation family. In a girl with early-onset epileptic encephalopathy, we identified the de novo GRIN2A mutation c.1845C>A predicting the amino acid substitution p.N615K. Analysis of NR1-NR2A(N615K) (NR2A subunit with the p.N615K alteration) receptor currents revealed a loss of the Mg²(+) block and a decrease in Ca²(+) permeability. Our findings suggest that disturbances in the neuronal electrophysiological balance during development result in variable neurological phenotypes depending on which NR2 subunit of NMDA receptors is affected.
Collapse
|
36
|
Abstract
During the past five years, copy number variation (CNV) has emerged as a highly prevalent form of genomic variation, bridging the interval between long-recognised microscopic chromosomal alterations and single-nucleotide changes. These genomic segmental differences among humans reflect the dynamic nature of genomes, and account for both normal variations among us and variations that predispose to conditions of medical consequence. Here, we place CNVs into their historical and medical contexts, focusing on how these variations can be recognised, documented, characterised and interpreted in clinical diagnostics. We also discuss how they can cause disease or influence adaptation to an environment. Various clinical exemplars are drawn out to illustrate salient characteristics and residual enigmas of CNVs, particularly the complexity of the data and information associated with CNVs relative to that of single-nucleotide variation. The potential is immense for CNVs to explain and predict disorders and traits that have long resisted understanding. However, creative solutions are needed to manage the sudden and overwhelming burden of expectation for laboratories and clinicians to assay and interpret these complex genomic variations as awareness permeates medical practice. Challenges remain for understanding the relationship between genomic changes and the phenotypes that might be predicted and prevented by such knowledge.
Collapse
|
37
|
Farini A, Razini P, Erratico S, Torrente Y, Meregalli M. Cell based therapy for Duchenne muscular dystrophy. J Cell Physiol 2009; 221:526-34. [PMID: 19688776 DOI: 10.1002/jcp.21895] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mutations in the dystrophin gene cause an X-linked genetic disorder: Duchenne muscular dystrophy (DMD). Stem cell therapy is an attractive method to treat DMD because a small number of cells are required to obtain a therapeutic effect. Here, we discussed about multiple types of myogenic stem cells and their possible use to treat DMD. The identification of a stem cell population providing efficient muscle regeneration is critical for the progression of cell therapy for DMD. We speculated that the most promising possibility for the treatment of DMD is a combination of different approaches, such as gene and stem cell therapy.
Collapse
Affiliation(s)
- Andrea Farini
- Stem Cell Laboratory, Department of Neurological Science, Centro Dino Ferrari, University of Milan, Fondazione IRCCS Policlinico Mangiagalli Regina Elena, Italy
| | | | | | | | | |
Collapse
|
38
|
X;7 Translocation in an Indian Woman with Hypergonadotropic Amenorrhea—A Case Report. Genet Test Mol Biomarkers 2009; 13:533-6. [DOI: 10.1089/gtmb.2009.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
39
|
Bhoj EJ, Romeo S, Baroni MG, Bartov G, Schultz RA, Zinn AR. MODY-like diabetes associated with an apparently balanced translocation: possible involvement of MPP7 gene and cell polarity in the pathogenesis of diabetes. Mol Cytogenet 2009; 2:5. [PMID: 19216786 PMCID: PMC2646739 DOI: 10.1186/1755-8166-2-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Accepted: 02/13/2009] [Indexed: 11/10/2022] Open
Abstract
Background Characterization of disease-associated balanced translocations has led to the discovery of genes responsible for many disorders, including syndromes that include various forms of diabetes mellitus. We studied a man with unexplained maturity onset diabetes of the young (MODY)-like diabetes and an apparently balanced translocation [46,XY,t(7;10)(q22;p12)] and sought to identify a novel diabetes locus by characterizing the translocation breakpoints. Results Mutations in coding exons and splice sites of known MODY genes were first ruled out by PCR amplification and DNA sequencing. Fluorescent in situ hybridization (FISH) studies demonstrated that the translocation did not disrupt two known diabetes-related genes on 10p12. The translocation breakpoints were further mapped to high resolution using FISH and somatic cell hybrids and the junctions PCR-amplified and sequenced. The translocation did not disrupt any annotated transcription unit. However, the chromosome 10 breakpoint was 220 kilobases 5' to the Membrane Protein, Palmitoylated 7 (MPP7) gene, which encodes a protein required for proper cell polarity. This biological function is shared by HNF4A, a known MODY gene. Databases show MPP7 is highly expressed in mouse pancreas and is expressed in human islets. The translocation did not appear to alter lymphoblastoid expression of MPP7 or other genes near the breakpoints. Conclusion The balanced translocation and MODY-like diabetes in the proband could be coincidental. Alternatively, the translocation may cause islet cell dysfunction by altering MPP7 expression in a subtle or tissue-specific fashion. The potential roles of MPP7 mutations in diabetes and perturbed islet cell polarity in insulin secretion warrant further study.
Collapse
Affiliation(s)
- Elizabeth J Bhoj
- McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Kulkarni S, Nagarajan P, Wall J, Donovan DJ, Donell RL, Ligon AH, Venkatachalam S, Quade BJ. Disruption of chromodomain helicase DNA binding protein 2 (CHD2) causes scoliosis. Am J Med Genet A 2008; 146A:1117-27. [PMID: 18386809 DOI: 10.1002/ajmg.a.32178] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Herein we characterize an apparently balanced de novo translocation, t(X;15)(p22.2;q26.1)dn, in a female patient with scoliosis, hirsutism, learning problems, and developmental delay (DGAP025). Other clinical findings include a high-arched palate, 2-3 syndactyly of the toes, and mildly elevated serum testosterone. No known or predicted genes are disrupted by the Xp22.2 breakpoint. The 15q26.1 breakpoint disrupts chromodomain helicase DNA binding protein 2 (CHD2). Another member of the chromatin-remodeling gene family, CHD7, has been associated with a defined constellation of congenital anomalies known as coloboma, heart anomaly, choanal atresia, mental retardation, genital and ear anomalies syndrome (CHARGE) and idiopathic scoliosis. Monosomy of 15q26 also has been associated with a spectrum of congenital abnormalities and growth retardation that overlaps with those of DGAP025. To provide a biological correlate, we characterized a mutant mouse model with Chd2 disruption that is associated with embryonic and perinatal lethality. Expression analysis indicated that Chd2 is expressed in the heart, forebrain, extremities, facial and dorsal regions during specific times of embryonic development. Chd2(+/m) mice showed pronounced lordokyphosis, reduced body fat, postnatal runting, and growth retardation. These data suggest that haploinsufficiency for CHD2 could result in a complex of abnormal human phenotypes that includes scoliosis and possibly features similar to CHARGE syndrome.
Collapse
Affiliation(s)
- Shashikant Kulkarni
- Division of Women's and Perinatal Pathology and Clinical Cytogenetics Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Fantes JA, Boland E, Ramsay J, Donnai D, Splitt M, Goodship JA, Stewart H, Whiteford M, Gautier P, Harewood L, Holloway S, Sharkey F, Maher E, van Heyningen V, Clayton-Smith J, Fitzpatrick DR, Black GCM. FISH mapping of de novo apparently balanced chromosome rearrangements identifies characteristics associated with phenotypic abnormality. Am J Hum Genet 2008; 82:916-26. [PMID: 18374296 PMCID: PMC2491339 DOI: 10.1016/j.ajhg.2008.02.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 02/03/2008] [Accepted: 02/05/2008] [Indexed: 11/16/2022] Open
Abstract
We report fluorescence in situ hybridization (FISH) mapping of 152, mostly de novo, apparently balanced chromosomal rearrangement (ABCR) breakpoints in 76 individuals, 30 of whom had no obvious phenotypic abnormality (control group) and 46 of whom had an associated disease (case group). The aim of this study was to identify breakpoint characteristics that could discriminate between these groups and which might be of predictive value in de novo ABCR (DN-ABCR) cases detected antenatally. We found no difference in the proportion of breakpoints that interrupted a gene, although in three cases, direct interruption or deletion of known autosomal-dominant or X-linked recessive Mendelian disease genes was diagnostic. The only significant predictor of phenotypic abnormality in the group as a whole was the localization of one or both breakpoints to an R-positive (G-negative) band with estimated predictive values of 0.69 (95% CL 0.54-0.81) and 0.90 (95% CL 0.60-0.98), respectively. R-positive bands are known to contain more genes and have a higher guanine-cytosine (GC) content than do G-positive (R-negative) bands; however, whether a gene was interrupted by the breakpoint or the GC content in the 200 kB around the breakpoint had no discriminant ability. Our results suggest that the large-scale genomic context of the breakpoint has prognostic utility and that the pathological mechanism of mapping to an R-band cannot be accounted for by direct gene inactivation.
Collapse
Affiliation(s)
- J A Fantes
- Medical and Developmental Genetics Section, Medical Research Council (MRC), Human Genetics Unit, Edinburgh EH4 2XU, Scotland, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Schoeffler M, Wallet F, Robert M, Tramoni G, Workineh S, Viale J, Duperret S. Élévation de troponine I à coronarographie normale chez un patient porteur d’une myopathie de Duchenne. ACTA ACUST UNITED AC 2008; 27:345-7. [DOI: 10.1016/j.annfar.2008.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2007] [Accepted: 01/08/2008] [Indexed: 11/26/2022]
|
43
|
Higgins AW, Alkuraya FS, Bosco AF, Brown KK, Bruns GA, Donovan DJ, Eisenman R, Fan Y, Farra CG, Ferguson HL, Gusella JF, Harris DJ, Herrick SR, Kelly C, Kim HG, Kishikawa S, Korf BR, Kulkarni S, Lally E, Leach NT, Lemyre E, Lewis J, Ligon AH, Lu W, Maas RL, MacDonald ME, Moore SD, Peters RE, Quade BJ, Quintero-Rivera F, Saadi I, Shen Y, Shendure J, Williamson RE, Morton CC. Characterization of apparently balanced chromosomal rearrangements from the developmental genome anatomy project. Am J Hum Genet 2008; 82:712-22. [PMID: 18319076 DOI: 10.1016/j.ajhg.2008.01.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 12/17/2007] [Accepted: 01/04/2008] [Indexed: 12/27/2022] Open
Abstract
Apparently balanced chromosomal rearrangements in individuals with major congenital anomalies represent natural experiments of gene disruption and dysregulation. These individuals can be studied to identify novel genes critical in human development and to annotate further the function of known genes. Identification and characterization of these genes is the goal of the Developmental Genome Anatomy Project (DGAP). DGAP is a multidisciplinary effort that leverages the recent advances resulting from the Human Genome Project to increase our understanding of birth defects and the process of human development. Clinically significant phenotypes of individuals enrolled in DGAP are varied and, in most cases, involve multiple organ systems. Study of these individuals' chromosomal rearrangements has resulted in the mapping of 77 breakpoints from 40 chromosomal rearrangements by FISH with BACs and fosmids, array CGH, Southern-blot hybridization, MLPA, RT-PCR, and suppression PCR. Eighteen chromosomal breakpoints have been cloned and sequenced. Unsuspected genomic imbalances and cryptic rearrangements were detected, but less frequently than has been reported previously. Chromosomal rearrangements, both balanced and unbalanced, in individuals with multiple congenital anomalies continue to be a valuable resource for gene discovery and annotation.
Collapse
|
44
|
Duboc D, Meune C, Pierre B, Wahbi K, Eymard B, Toutain A, Berard C, Vaksmann G, Weber S, Bécane HM. Perindopril preventive treatment on mortality in Duchenne muscular dystrophy: 10 years' follow-up. Am Heart J 2007; 154:596-602. [PMID: 17719312 DOI: 10.1016/j.ahj.2007.05.014] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Accepted: 05/14/2007] [Indexed: 12/16/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD), an X-linked disorder due to lack of dystrophin, is associated with muscle weakness and myocardial dysfunction. Although preliminary data support the efficacy of angiotensin-converting enzyme inhibitors on left ventricular (LV) function, our aim was to examine the long-term impact of a preventive treatment with perindopril on mortality in children with DMD. METHODS Patients with DMD between the ages of 9.5 and 13 years presenting with normal LV ejection fraction were included in this prospective study. They were randomly assigned for 3 years to perindopril, 2 to 4 mg (group 1), or placebo (group 2) in a double-blind protocol, followed by open-label treatment with perindopril for up to 10 years. Survival rate at 10 years in each group is reported. RESULTS There were 28 patients assigned to group 1 and 29 to group 2. Baseline characteristics were similar in both groups. At the end of the 10 years' follow-up period, survival status was available for all included patients: 26 (92.9%) of 28 patients in group 1 were alive at 10 years versus 19 (65.5%) of 29 in group 2 (P = .02). Kaplan-Meier cumulative survival was significantly lower in group 2 than in group 1 (P = .013). CONCLUSION Early initiation of treatment with perindopril is associated with a lower mortality in patients with DMD with normal LV ejection fraction at study entry.
Collapse
Affiliation(s)
- Denis Duboc
- Department of Cardiology, Cochin Hospital, APHP, Paris V René Descartes University, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Duboc D, Meune C, Pierre B, Wahbi K, Eymard B, Toutain A, Berard C, Vaksmann G, Bécane HM. Perindopril preserves left ventricular function in X-linked Duchenne muscular dystrophy. Eur Heart J Suppl 2007. [DOI: 10.1093/eurheartj/sum036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
46
|
Silva MC, Meira ZMA, Gurgel Giannetti J, da Silva MM, Campos AFO, Barbosa MDM, Starling Filho GM, Ferreira RDA, Zatz M, Rochitte CE. Myocardial delayed enhancement by magnetic resonance imaging in patients with muscular dystrophy. J Am Coll Cardiol 2007; 49:1874-9. [PMID: 17481447 DOI: 10.1016/j.jacc.2006.10.078] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Revised: 09/21/2006] [Accepted: 10/16/2006] [Indexed: 12/17/2022]
Abstract
OBJECTIVES This study sought to analyze whether cardiovascular magnetic resonance (CMR) can detect and quantify myocardial damage in the early stages of cardiomyopathy in muscular dystrophies (MD). BACKGROUND Muscular dystrophy is a genetic disease that involves skeletal and cardiac tissues of humans. Cardiomyopathy is common, and death secondary to cardiac or respiratory diseases occurs early in life. Cardiovascular magnetic resonance is a reliable method for assessing global and regional cardiac function, allowing also for the detection of myocardial fibrosis (MF). METHODS Ten patients with Duchenne or Becker dystrophies were studied by CMR. Physical examination, Chagas disease serological tests, electrocardiogram, chest radiograph, total creatine kinase, and Doppler echocardiogram were also obtained in all patients. RESULTS Patients with MF had a lower ejection fraction than those without. Myocardial fibrosis (midwall and/or subepicardial) was observed in 7 of the 10 patients, and the lateral wall was the most commonly involved segment. There was moderate correlation between segmental MF and dysfunction. CONCLUSIONS Cardiovascular magnetic resonance can identify MF and may be useful for detecting the early stages of cardiomyopathy in MD. Future work will be needed to evaluate whether CMR can influence cardiomyopathy and outcomes.
Collapse
|
47
|
Suzuki N, Miyagoe-Suzuki Y, Takeda S. Gene therapy for Duchenne muscular dystrophy. FUTURE NEUROLOGY 2007. [DOI: 10.2217/14796708.2.1.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gene therapy has great potential to treat Duchenne muscular dystrophy. Among many proposed strategies to deliver a therapeutic gene to muscle, recombinant adeno-associated virus-mediated gene transfer is the most promising. The recent isolation of new adeno-associated virus serotypes from human and nonhuman primates provides the opportunity to develop vectors that can achieve the long-term expression of a therapeutic gene in muscles of the entire body without detrimental effects. To translate the results from small animal models to clinical trials in humans, further work using larger animal models, such as dystrophic dogs or nonhuman primates, is required. This review also discusses recent progress in other gene transfer-related therapeutic approaches, including targeted exon skipping and gene correction.
Collapse
Affiliation(s)
- Naoki Suzuki
- National Institute of Neuroscience, Department of Molecular Therapy, National Center of Neurology & Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Yuko Miyagoe-Suzuki
- National Institute of Neuroscience, Department of Molecular Therapy, National Center of Neurology & Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Shin’ichi Takeda
- National Institute of Neuroscience, Department of Molecular Therapy, National Center of Neurology & Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187–8502, Japan
| |
Collapse
|
48
|
White SJ, den Dunnen JT. Copy number variation in the genome; the human DMD gene as an example. Cytogenet Genome Res 2006; 115:240-6. [PMID: 17124406 DOI: 10.1159/000095920] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Accepted: 05/15/2006] [Indexed: 11/19/2022] Open
Abstract
Recent developments have yielded new technologies that have greatly simplified the detection of deletions and duplications, i.e., copy number variants (CNVs). These technologies can be used to screen for CNVs in and around specific genomic regions, as well as genome-wide. Several genome-wide studies have demonstrated that CNV in the human genome is widespread and may include millions of nucleotides. One of the questions that emerge is which sequences, structures and/or processes are involved in their generation. Using as an example the human DMD gene, mutations in which cause Duchenne and Becker muscular dystrophy, we review the current data, determine the deletion and duplication profile across the gene and summarize the information that has been collected regarding their origin. In addition we discuss the methods most frequently used for their detection, in particular MAPH and MLPA.
Collapse
Affiliation(s)
- S J White
- Human and Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
49
|
Price FD, Kuroda K, Rudnicki MA. Stem cell based therapies to treat muscular dystrophy. Biochim Biophys Acta Mol Basis Dis 2006; 1772:272-83. [PMID: 17034994 DOI: 10.1016/j.bbadis.2006.08.011] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 08/30/2006] [Indexed: 01/14/2023]
Abstract
Muscular dystrophies comprise a heterogeneous group of neuromuscular disorders, characterized by progressive muscle wasting, for which no satisfactory treatment exists. Multiple stem cell populations, both of adult or embryonic origin, display myogenic potential and have been assayed for their ability to correct the dystrophic phenotype. To date, many of these described methods have failed, underlying the need to identify the mechanisms controlling myogenic potential, homing of donor populations to the musculature, and avoidance of the immune response. Recent results focus on the fresh isolation of satellite cells and the use of multiple growth factors to promote mesangioblast migration, both of which promote muscle regeneration. Throughout this chapter, various stem cell based therapies will be introduced and evaluated based on their potential to treat muscular dystrophy in an effective and efficient manner.
Collapse
Affiliation(s)
- F D Price
- Molecular Medicine Program and Center for Stem Cell and Gene Therapy, Ottawa Health Research Institute, 501 Smyth Road, Ottawa, ON, Canada K1H 8L6
| | | | | |
Collapse
|
50
|
Johnson D, Morrison N, Grant L, Turner T, Fantes J, Connor JM, Murday V. Confirmation of CHD7 as a cause of CHARGE association identified by mapping a balanced chromosome translocation in affected monozygotic twins. J Med Genet 2006; 43:280-4. [PMID: 16118347 PMCID: PMC2563251 DOI: 10.1136/jmg.2005.032946] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Revised: 06/02/2005] [Accepted: 06/02/2005] [Indexed: 11/04/2022]
Abstract
BACKGROUND CHARGE syndrome has an estimated prevalence of 1/10,000. Most cases are sporadic which led to hypotheses of a non-genetic aetiology. However, there was also evidence for a genetic cause with reports of multiplex families with presumed autosomal dominant, possible autosomal recessive inheritance and concordant twin pairs. We identified a monozygotic twin pair with CHARGE syndrome and a de novo balanced chromosome rearrangement t(8;13)(q11.2;q22). METHODS Fluorescence in situ hybridisation was performed with BAC and PAC probes to characterise the translocation breakpoints. The breakpoint on chromosome 8 was further refined using 10 kb probes we designed and produced using sequence data for clone RP11 33I11, the Primer3 website, and a long range PCR kit. RESULTS BAC and PAC probe hybridisation redefined the breakpoints to 8q12.2 and 13q31.1. Probe RP11 33I11 spanned the breakpoint on chromosome 8. Using our 10 kb probes we demonstrated that the chromodomain gene CHD7 was disrupted by the translocation between exons 3 and 8. DISCUSSION Identifying that the translocation breakpoint in our patients occurred between exons 3 and 8 of CHD7 suggests that disruption of this gene is the cause of CHARGE syndrome in the twins and independently confirms the role of CHD7 in CHARGE syndrome.
Collapse
|