151
|
Tasca G, Pescatori M, Monforte M, Mirabella M, Iannaccone E, Frusciante R, Cubeddu T, Laschena F, Ottaviani P, Ricci E. Different molecular signatures in magnetic resonance imaging-staged facioscapulohumeral muscular dystrophy muscles. PLoS One 2012; 7:e38779. [PMID: 22719944 PMCID: PMC3374833 DOI: 10.1371/journal.pone.0038779] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 05/10/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common muscular dystrophies and is characterized by a non-conventional genetic mechanism activated by pathogenic D4Z4 repeat contractions. By muscle Magnetic Resonance Imaging (MRI) we observed that T2-short tau inversion recovery (T2-STIR) sequences identify two different conditions in which each muscle can be found before the irreversible dystrophic alteration, marked as T1-weighted sequence hyperintensity, takes place. We studied these conditions in order to obtain further information on the molecular mechanisms involved in the selective wasting of single muscles or muscle groups in this disease. METHODS Histopathology, gene expression profiling and real time PCR were performed on biopsies from FSHD muscles with different MRI pattern (T1-weighted normal/T2-STIR normal and T1-weighted normal/T2-STIR hyperintense). Data were compared with those from inflammatory myopathies, dysferlinopathies and normal controls. In order to validate obtained results, two additional FSHD samples with different MRI pattern were analyzed. RESULTS Myopathic and inflammatory changes characterized T2-STIR hyperintense FSHD muscles, at variance with T2-STIR normal muscles. These two states could be easily distinguished from each other by their transcriptional profile. The comparison between T2-STIR hyperintense FSHD muscles and inflammatory myopathy muscles showed peculiar changes, although many alterations were shared among these conditions. CONCLUSIONS At the single muscle level, different stages of the disease correspond to the two MRI patterns. T2-STIR hyperintense FSHD muscles are more similar to inflammatory myopathies than to T2-STIR normal FSHD muscles or other muscular dystrophies, and share with them upregulation of genes involved in innate and adaptive immunity. Our data suggest that selective inflammation, together with perturbation in biological processes such as neoangiogenesis, lipid metabolism and adipokine production, may contribute to the sequential bursts of muscle degeneration that involve individual muscles in an asynchronous manner in this disease.
Collapse
Affiliation(s)
| | - Mario Pescatori
- Department of Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
- Crosslinks BV, Rotterdam, The Netherlands
- Porto Conte Ricerche Srl, Tramariglio, Alghero (SS), Italy
| | - Mauro Monforte
- Institute of Neurology, Catholic University School of Medicine, Rome, Italy
| | | | | | - Roberto Frusciante
- Institute of Neurology, Catholic University School of Medicine, Rome, Italy
| | | | - Francesco Laschena
- Department of Radiology, Istituto Dermopatico dell’Immacolata IRCCS, Rome, Italy
| | | | - Enzo Ricci
- Institute of Neurology, Catholic University School of Medicine, Rome, Italy
- * E-mail:
| |
Collapse
|
152
|
Desguerre I, Arnold L, Vignaud A, Cuvellier S, Yacoub-youssef H, Gherardi RK, Chelly J, Chretien F, Mounier RÉ, Ferry A, Chazaud BÉ. A new model of experimental fibrosis in hindlimb skeletal muscle of adult mdx mouse mimicking muscular dystrophy. Muscle Nerve 2012; 45:803-14. [DOI: 10.1002/mus.23341] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
153
|
De Pasquale L, D'Amico A, Verardo M, Petrini S, Bertini E, De Benedetti F. Increased muscle expression of interleukin-17 in Duchenne muscular dystrophy. Neurology 2012; 78:1309-14. [PMID: 22496194 DOI: 10.1212/wnl.0b013e3182518302] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVES Duchenne muscular dystrophy (DMD) is a degenerative muscle wasting disease caused by mutations in the dystrophin gene. Dystrophic muscle is characterized by chronic inflammation, and inflammatory mediators could be promising targets for innovative therapeutic interventions. We analyzed muscle biopsy samples of DMD-affected children to characterize interleukin (IL)-17 and Forkhead box P3 (Foxp3) expression levels and to identify possible correlations with clinical status. METHODS Expression levels of IL-17, Foxp3, tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), IL-6, and transforming growth factor-β (TGF-β) were analyzed by real-time PCR in muscle biopsy samples from patients with DMD (n = 27) and juvenile dermatomyositis (JDM) (n = 8). Motor outcome of patients with DMD was evaluated by North Star Ambulatory Assessment score. RESULTS In DMD, we found higher levels of IL-17 and lower levels of Foxp3 mRNA compared with those for a typical inflammatory myopathy, JDM. Moreover, the IL-17/Foxp3 ratio was higher in DMD than in JDM biopsy samples. IL-17 mRNA levels appeared to be related to the expression levels of other proinflammatory cytokines (TNF-α and MCP-1) and significantly associated with clinical outcome of patients. CONCLUSIONS The association of IL-17 expression with levels of other inflammatory cytokines and with the clinical course of DMD suggests a possible pathogenic role of IL-17.
Collapse
Affiliation(s)
- L De Pasquale
- Division of Rheumatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | | | | | | | | |
Collapse
|
154
|
Kemaladewi DU, ‘t Hoen PA, ten Dijke P, van Ommen GJ, Hoogaars WM. TGF-β signaling in Duchenne muscular dystrophy. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The TGF-β protein family consists of secreted multifunctional cytokines that control diverse processes, such as cell growth and differentiation. Aberrant expression and downstream signaling of these growth factors have been associated with multiple diseases, including muscle wasting disorders, such as Duchenne muscular dystrophy. In this review we discuss recent advances in understanding the role of TGF-β family members during normal skeletal muscle biology/regeneration and their role in muscle pathology, with a special focus on Duchenne muscular dystrophy. In addition, we will highlight progress in the development of potential therapeutics for Duchenne muscular dystrophy based on intervention of TGF-β signaling.
Collapse
Affiliation(s)
- Dwi U Kemaladewi
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
- Department of Molecular & Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Peter A ‘t Hoen
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Peter ten Dijke
- Department of Molecular & Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Gert Jan van Ommen
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| | - Willem M Hoogaars
- Department of Human Genetics, Leiden University Medical Center, Postzone S4-P, PO Box 9600 2300RC Leiden, The Netherlands
| |
Collapse
|
155
|
Kotelnikova E, Shkrob MA, Pyatnitskiy MA, Ferlini A, Daraselia N. Novel approach to meta-analysis of microarray datasets reveals muscle remodeling-related drug targets and biomarkers in Duchenne muscular dystrophy. PLoS Comput Biol 2012; 8:e1002365. [PMID: 22319435 PMCID: PMC3271016 DOI: 10.1371/journal.pcbi.1002365] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 12/15/2011] [Indexed: 12/24/2022] Open
Abstract
Elucidation of new biomarkers and potential drug targets from high-throughput profiling data is a challenging task due to a limited number of available biological samples and questionable reproducibility of differential changes in cross-dataset comparisons. In this paper we propose a novel computational approach for drug and biomarkers discovery using comprehensive analysis of multiple expression profiling datasets. The new method relies on aggregation of individual profiling experiments combined with leave-one-dataset-out validation approach. Aggregated datasets were studied using Sub-Network Enrichment Analysis algorithm (SNEA) to find consistent statistically significant key regulators within the global literature-extracted expression regulation network. These regulators were linked to the consistent differentially expressed genes. We have applied our approach to several publicly available human muscle gene expression profiling datasets related to Duchenne muscular dystrophy (DMD). In order to detect both enhanced and repressed processes we considered up- and down-regulated genes separately. Applying the proposed approach to the regulators search we discovered the disturbance in the activity of several muscle-related transcription factors (e.g. MYOG and MYOD1), regulators of inflammation, regeneration, and fibrosis. Almost all SNEA-derived regulators of down-regulated genes (e.g. AMPK, TORC2, PPARGC1A) correspond to a single common pathway important for fast-to-slow twitch fiber type transition. We hypothesize that this process can affect the severity of DMD symptoms, making corresponding regulators and downstream genes valuable candidates for being potential drug targets and exploratory biomarkers. Comparison of gene expression in diseased and normal tissue is a powerful tool of studying processes involved in pathogenesis and searching for potential drug targets and biomarkers of the disease's progression and treatment outcome. We have developed a novel approach for systematic knowledge-driven analysis of gene expression profiling data, which can suggest the underlying cause of the observed differential expression by identifying which expression regulators might be involved. These regulators can not only be the promising subjects of further investigation, but also potential drug targets, as normalization of their activity might alleviate some of the disease's symptoms. The targets downstream of suggested regulators can be proposed as exploratory biomarkers in disease treatment and prognosis. We used our approach to analyze public gene expression datasets of Duchenne muscular dystrophy – a progressive inherited disease in males. Some of the regulators and biomarkers that we found were already investigated in the context of DMD, while some of them were not yet studied and may be of interest for biological and clinical studies.
Collapse
|
156
|
Selsby JT, Morine KJ, Pendrak K, Barton ER, Sweeney HL. Rescue of dystrophic skeletal muscle by PGC-1α involves a fast to slow fiber type shift in the mdx mouse. PLoS One 2012; 7:e30063. [PMID: 22253880 PMCID: PMC3256197 DOI: 10.1371/journal.pone.0030063] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 12/12/2011] [Indexed: 11/19/2022] Open
Abstract
Increased utrophin expression is known to reduce pathology in dystrophin-deficient skeletal muscles. Transgenic over-expression of PGC-1α has been shown to increase levels of utrophin mRNA and improve the histology of mdx muscles. Other reports have shown that PGC-1α signaling can lead to increased oxidative capacity and a fast to slow fiber type shift. Given that it has been shown that slow fibers produce and maintain more utrophin than fast skeletal muscle fibers, we hypothesized that over-expression of PGC-1α in post-natal mdx mice would increase utrophin levels via a fiber type shift, resulting in more slow, oxidative fibers that are also more resistant to contraction-induced damage. To test this hypothesis, neonatal mdx mice were injected with recombinant adeno-associated virus (AAV) driving expression of PGC-1α. PGC-1α over-expression resulted in increased utrophin and type I myosin heavy chain expression as well as elevated mitochondrial protein expression. Muscles were shown to be more resistant to contraction-induced damage and more fatigue resistant. Sirt-1 was increased while p38 activation and NRF-1 were reduced in PGC-1α over-expressing muscle when compared to control. We also evaluated if the use a pharmacological PGC-1α pathway activator, resveratrol, could drive the same physiological changes. Resveratrol administration (100 mg/kg/day) resulted in improved fatigue resistance, but did not achieve significant increases in utrophin expression. These data suggest that the PGC-1α pathway is a potential target for therapeutic intervention in dystrophic skeletal muscle.
Collapse
Affiliation(s)
- Joshua T. Selsby
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Animal Science, College of Agriculture and Life Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Kevin J. Morine
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Klara Pendrak
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elisabeth R. Barton
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - H. Lee Sweeney
- Department of Physiology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
157
|
Soh D, Dong D, Guo Y, Wong L. Finding consistent disease subnetworks across microarray datasets. BMC Bioinformatics 2011; 12 Suppl 13:S15. [PMID: 22372958 PMCID: PMC3278831 DOI: 10.1186/1471-2105-12-s13-s15] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background While contemporary methods of microarray analysis are excellent tools for studying individual microarray datasets, they have a tendency to produce different results from different datasets of the same disease. We aim to solve this reproducibility problem by introducing a technique (SNet). SNet provides both quantitative and descriptive analysis of microarray datasets by identifying specific connected portions of pathways that are significant. We term such portions within pathways as “subnetworks”. Results We tested SNet on independent datasets of several diseases, including childhood ALL, DMD and lung cancer. For each of these diseases, we obtained two independent microarray datasets produced by distinct labs on distinct platforms. In each case, our technique consistently produced almost the same list of significant nontrivial subnetworks from two independent sets of microarray data. The gene-level agreement of these significant subnetworks was between 51.18% to 93.01%. In contrast, when the same pairs of microarray datasets were analysed using GSEA, t-test and SAM, this percentage fell between 2.38% to 28.90% for GSEA, 49.60% tp 73.01% for t-test, and 49.96% to 81.25% for SAM. Furthermore, the genes selected using these existing methods did not form subnetworks of substantial size. Thus it is more probable that the subnetworks selected by our technique can provide the researcher with more descriptive information on the portions of the pathway actually affected by the disease. Conclusions These results clearly demonstrate that our technique generates significant subnetworks and genes that are more consistent and reproducible across datasets compared to the other popular methods available (GSEA, t-test and SAM). The large size of subnetworks which we generate indicates that they are generally more biologically significant (less likely to be spurious). In addition, we have chosen two sample subnetworks and validated them with references from biological literature. This shows that our algorithm is capable of generating descriptive biologically conclusions.
Collapse
Affiliation(s)
- Donny Soh
- National University of Singapore, 13 Computing Drive, Singapore 117417.
| | | | | | | |
Collapse
|
158
|
LIU ZIYING, PHAN SIEU, FAMILI FAZEL, PAN YOULIAN, LENFERINK ANNEEG, CANTIN CHRISTIANE, COLLINS CATHERINE, O'CONNOR-MCCOURT MAUREEND. A MULTI-STRATEGY APPROACH TO INFORMATIVE GENE IDENTIFICATION FROM GENE EXPRESSION DATA. J Bioinform Comput Biol 2011; 8:19-38. [DOI: 10.1142/s0219720010004495] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 10/19/2009] [Accepted: 10/19/2009] [Indexed: 11/18/2022]
Abstract
An unsupervised multi-strategy approach has been developed to identify informative genes from high throughput genomic data. Several statistical methods have been used in the field to identify differentially expressed genes. Since different methods generate different lists of genes, it is very challenging to determine the most reliable gene list and the appropriate method. This paper presents a multi-strategy method, in which a combination of several data analysis techniques are applied to a given dataset and a confidence measure is established to select genes from the gene lists generated by these techniques to form the core of our final selection. The remainder of the genes that form the peripheral region are subject to exclusion or inclusion into the final selection. This paper demonstrates this methodology through its application to an in-house cancer genomics dataset and a public dataset. The results indicate that our method provides more reliable list of genes, which are validated using biological knowledge, biological experiments, and literature search. We further evaluated our multi-strategy method by consolidating two pairs of independent datasets, each pair is for the same disease, but generated by different labs using different platforms. The results showed that our method has produced far better results.
Collapse
Affiliation(s)
- ZIYING LIU
- Institute for Information Technology, National Research Council Canada, Ottawa, Ontario, K1A 0R6, Canada
| | - SIEU PHAN
- Institute for Information Technology, National Research Council Canada, Ottawa, Ontario, K1A 0R6, Canada
| | - FAZEL FAMILI
- Institute for Information Technology, National Research Council Canada, Ottawa, Ontario, K1A 0R6, Canada
| | - YOULIAN PAN
- Institute for Information Technology, National Research Council Canada, Ottawa, Ontario, K1A 0R6, Canada
| | - ANNE E. G. LENFERINK
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec, H4P 2R2, Canada
| | - CHRISTIANE CANTIN
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec, H4P 2R2, Canada
| | - CATHERINE COLLINS
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec, H4P 2R2, Canada
| | | |
Collapse
|
159
|
Cirak S, Arechavala-Gomeza V, Guglieri M, Feng L, Torelli S, Anthony K, Abbs S, Garralda ME, Bourke J, Wells DJ, Dickson G, Wood MJA, Wilton SD, Straub V, Kole R, Shrewsbury SB, Sewry C, Morgan JE, Bushby K, Muntoni F. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet 2011; 378:595-605. [PMID: 21784508 PMCID: PMC3156980 DOI: 10.1016/s0140-6736(11)60756-3] [Citation(s) in RCA: 681] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND We report clinical safety and biochemical efficacy from a dose-ranging study of intravenously administered AVI-4658 phosphorodiamidate morpholino oligomer (PMO) in patients with Duchenne muscular dystrophy. METHOD We undertook an open-label, phase 2, dose-escalation study (0·5, 1·0, 2·0, 4·0, 10·0, and 20·0 mg/kg bodyweight) in ambulant patients with Duchenne muscular dystrophy aged 5-15 years with amenable deletions in DMD. Participants had a muscle biopsy before starting treatment and after 12 weekly intravenous infusions of AVI-4658. The primary study objective was to assess safety and tolerability of AVI-4658. The secondary objectives were pharmacokinetic properties and the ability of AVI-4658 to induce exon 51 skipping and dystrophin restoration by RT-PCR, immunohistochemistry, and immunoblotting. The study is registered, number NCT00844597. FINDINGS 19 patients took part in the study. AVI-4658 was well tolerated with no drug-related serious adverse events. AVI-4658 induced exon 51 skipping in all cohorts and new dystrophin protein expression in a significant dose-dependent (p=0·0203), but variable, manner in boys from cohort 3 (dose 2 mg/kg) onwards. Seven patients responded to treatment, in whom mean dystrophin fluorescence intensity increased from 8·9% (95% CI 7·1-10·6) to 16·4% (10·8-22·0) of normal control after treatment (p=0·0287). The three patients with the greatest responses to treatment had 21%, 15%, and 55% dystrophin-positive fibres after treatment and these findings were confirmed with western blot, which showed an increase after treatment of protein levels from 2% to 18%, from 0·9% to 17%, and from 0% to 7·7% of normal muscle, respectively. The dystrophin-associated proteins α-sarcoglycan and neuronal nitric oxide synthase were also restored at the sarcolemma. Analysis of the inflammatory infiltrate indicated a reduction of cytotoxic T cells in the post-treatment muscle biopsies in the two high-dose cohorts. INTERPRETATION The safety and biochemical efficacy that we present show the potential of AVI-4658 to become a disease-modifying drug for Duchenne muscular dystrophy. FUNDING UK Medical Research Council; AVI BioPharma.
Collapse
Affiliation(s)
- Sebahattin Cirak
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | | | - Michela Guglieri
- Institute of Human Genetics, Newcastle University, Newcastle, UK
| | - Lucy Feng
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Silvia Torelli
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Karen Anthony
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Stephen Abbs
- DNA Laboratory Genetics Centre, Guy's Hospital, London, UK
| | - Maria Elena Garralda
- Academic Unit of Child and Adolescent Psychiatry, Imperial College St Mary's Campus, London, UK
| | - John Bourke
- Institute of Human Genetics, Newcastle University, Newcastle, UK
| | | | | | - Matthew JA Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Steve D Wilton
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Perth, WA, Australia
| | - Volker Straub
- Institute of Human Genetics, Newcastle University, Newcastle, UK
| | | | | | - Caroline Sewry
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
- Centre for Inherited Neuromuscular Disorders, RJAH Orthopaedic Hospital, Oswestry, UK
| | - Jennifer E Morgan
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Kate Bushby
- Institute of Human Genetics, Newcastle University, Newcastle, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| |
Collapse
|
160
|
Nadarajah VD, van Putten M, Chaouch A, Garrood P, Straub V, Lochmüller H, Ginjaar HB, Aartsma-Rus AM, van Ommen GJB, den Dunnen JT, 't Hoen PAC. Serum matrix metalloproteinase-9 (MMP-9) as a biomarker for monitoring disease progression in Duchenne muscular dystrophy (DMD). Neuromuscul Disord 2011; 21:569-78. [PMID: 21724396 DOI: 10.1016/j.nmd.2011.05.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 05/24/2011] [Accepted: 05/27/2011] [Indexed: 11/19/2022]
Abstract
To identify serum biomarkers that allow monitoring of disease progression and treatment effects in Duchenne muscular dystrophy (DMD) patients, levels of matrix metalloproteinase-9 (MMP-9), tissue inhibitors of metalloproteinase-1 (TIMP-1) and osteopontin (OPN) were determined in 63 DMD patients on corticosteroid therapy. These proteins were selected for their role in the pathogenesis of muscular dystrophy. Levels of MMP-9 and TIMP-1 were significantly higher in sera of DMD patients compared to healthy controls, whereas the OPN levels showed no significant difference. MMP-9 levels were also observed to be significantly higher in older, nonambulant patients, compared to ambulant patients. Longitudinal data from a smaller cohort of DMD patients followed up for over 4years showed that MMP-9, but not TIMP-1 increased significantly with age. Hence, MMP-9 is a potential DMD biomarker for disease progression. Future studies have to confirm whether serum MMP-9 levels can be used to monitor therapeutic response.
Collapse
Affiliation(s)
- V D Nadarajah
- Department of Human Genetics, Leiden University Medical Centre (LUMC), RC, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Ahmad N, Welch I, Grange R, Hadway J, Dhanvantari S, Hill D, Lee TY, Hoffman LM. Use of imaging biomarkers to assess perfusion and glucose metabolism in the skeletal muscle of dystrophic mice. BMC Musculoskelet Disord 2011; 12:127. [PMID: 21639930 PMCID: PMC3141608 DOI: 10.1186/1471-2474-12-127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 06/04/2011] [Indexed: 11/10/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease that affects 1 in 3500 boys. The disease is characterized by progressive muscle degeneration that results from mutations in or loss of the cytoskeletal protein, dystrophin, from the glycoprotein membrane complex, thus increasing the susceptibility of contractile muscle to injury. To date, disease progression is typically assessed using invasive techniques such as muscle biopsies, and while there are recent reports of the use of magnetic resonance, ultrasound and optical imaging technologies to address the issue of disease progression and monitoring therapeutic intervention in dystrophic mice, our study aims to validate the use of imaging biomarkers (muscle perfusion and metabolism) in a longitudinal assessment of skeletal muscle degeneration/regeneration in two murine models of muscular dystrophy. Methods Wild-type (w.t.) and dystrophic mice (weakly-affected mdx mice that are characterized by a point mutation in dystrophin; severely-affected mdx:utrn-/- (udx) mice that lack functional dystrophin and are null for utrophin) were exercised three times a week for 30 minutes. To follow the progression of DMD, accumulation of 18 F-FDG, a measure of glucose metabolism, in both wild-type and affected mice was measured with a small animal PET scanner (GE eXplore Vista). To assess changes in blood flow and blood volume in the hind limb skeletal muscle, mice were injected intravenously with a CT contrast agent, and imaged with a small animal CT scanner (GE eXplore Ultra). Results In hind limb skeletal muscle of both weakly-affected mdx mice and in severely-affected udx mice, we demonstrate an early, transient increase in both 18F-FDG uptake, and in blood flow and blood volume. Histological analysis of H&E-stained tissue collected from parallel littermates demonstrates the presence of both inflammatory infiltrate and centrally-located nuclei, a classic hallmark of myofibrillar regeneration. In both groups of affected mice, the early transient response was succeeded by a progressive decline in muscle perfusion and metabolism; this was also evidenced histologically. Conclusions The present study demonstrates the utility of non-invasive imaging biomarkers in characterizing muscle degeneration/regeneration in murine models of DMD. These techniques may now provide a promising alternative for assessing both disease progression and the efficacy of new therapeutic treatments in patients.
Collapse
Affiliation(s)
- Nabeel Ahmad
- Imaging Program, Lawson Health Research Institute, 268 Grosvenor St., London N6A4V2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
162
|
Lawler JM. Exacerbation of pathology by oxidative stress in respiratory and locomotor muscles with Duchenne muscular dystrophy. J Physiol 2011; 589:2161-70. [PMID: 21486793 PMCID: PMC3098695 DOI: 10.1113/jphysiol.2011.207456] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 03/02/2011] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most devastating type of muscular dystrophy, leading to progressive weakness of respiratory (e.g. diaphragm) and locomotor muscles (e.g. gastrocnemius). DMD is caused by X-linked defects in the gene that encodes for dystrophin, a key scaffolding protein of the dystroglycan complex (DCG) within the sarcolemmal cytoskeleton. As a result of a compromised dystroglycan complex, mechanical integrity is impaired and important signalling proteins (e.g. nNOS, caveolin-3) and pathways are disrupted. Disruption of the dystroglycan complex leads to high susceptibility to injury with repeated, eccentric contractions as well as inflammation, resulting in significant damage and necrosis. Chronic damage and repair cycling leads to fibrosis and weakness. While the link between inflammation with damage and weakness in the DMD diaphragm is unresolved, elevated oxidative stress may contribute to damage, weakness and possibly fibrosis. While utilization of non-specific antioxidant interventions has yielded inconsistent results, recent data suggest that NAD(P)H oxidase could play a pivotal role in elevating oxidative stress via integrated changes in caveolin-3 and stretch-activated channels (SACs). Oxidative stress may act as an amplifier, exacerbating disruption of the dystroglycan complex, upregulation of the inflammatory transcription factor NF-B, and thus functional impairment of force-generating capacity.
Collapse
Affiliation(s)
- John M Lawler
- Department of Health and Kinesiology, Texas A&M University, College Station, TX 77843-4243, USA.
| |
Collapse
|
163
|
Guevel L, Lavoie JR, Perez-Iratxeta C, Rouger K, Dubreil L, Feron M, Talon S, Brand M, Megeney LA. Quantitative proteomic analysis of dystrophic dog muscle. J Proteome Res 2011; 10:2465-78. [PMID: 21410286 DOI: 10.1021/pr2001385] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by null mutations in the dystrophin gene, leading to progressive and unrelenting muscle loss. Although the genetic basis of DMD is well resolved, the cellular mechanisms associated with the physiopathology remain largely unknown. Increasing evidence suggests that secondary mechanisms, as the alteration of key signaling pathways, may play an important role. In order to identify reliable biomarkers and potential therapeutic targets, and taking advantage of the clinically relevant Golden Retriever Muscular Dystrophy (GRMD) dog model, a proteomic study was performed. Isotope-coded affinity tag (ICAT) profiling was used to compile quantitative changes in protein expression profiles of the vastus lateralis muscles of 4-month old GRMD vs healthy dogs. Interestingly, the set of under-expressed proteins detected appeared primarily composed of metabolic proteins, many of which have been shown to be regulated by the transcriptional peroxisome proliferator-activated receptor-gamma co-activator 1 alpha (PGC-1α). Subsequently, we were able to showed that PGC1-α expression is dramatically reduced in GRMD compared to healthy muscle. Collectively, these results provide novel insights into the molecular pathology of the clinically relevant animal model of DMD, and indicate that defective energy metabolism is a central hallmark of the disease in the canine model.
Collapse
Affiliation(s)
- Laetitia Guevel
- CNRS UMR6204, Faculté des Sciences et des Techniques, F-44322 Nantes Cedex 3, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Baron D, Dubois E, Bihouée A, Teusan R, Steenman M, Jourdon P, Magot A, Péréon Y, Veitia R, Savagner F, Ramstein G, Houlgatte R. Meta-analysis of muscle transcriptome data using the MADMuscle database reveals biologically relevant gene patterns. BMC Genomics 2011; 12:113. [PMID: 21324190 PMCID: PMC3049149 DOI: 10.1186/1471-2164-12-113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 02/16/2011] [Indexed: 12/12/2022] Open
Abstract
Background DNA microarray technology has had a great impact on muscle research and microarray gene expression data has been widely used to identify gene signatures characteristic of the studied conditions. With the rapid accumulation of muscle microarray data, it is of great interest to understand how to compare and combine data across multiple studies. Meta-analysis of transcriptome data is a valuable method to achieve it. It enables to highlight conserved gene signatures between multiple independent studies. However, using it is made difficult by the diversity of the available data: different microarray platforms, different gene nomenclature, different species studied, etc. Description We have developed a system tool dedicated to muscle transcriptome data. This system comprises a collection of microarray data as well as a query tool. This latter allows the user to extract similar clusters of co-expressed genes from the database, using an input gene list. Common and relevant gene signatures can thus be searched more easily. The dedicated database consists in a large compendium of public data (more than 500 data sets) related to muscle (skeletal and heart). These studies included seven different animal species from invertebrates (Drosophila melanogaster, Caenorhabditis elegans) and vertebrates (Homo sapiens, Mus musculus, Rattus norvegicus, Canis familiaris, Gallus gallus). After a renormalization step, clusters of co-expressed genes were identified in each dataset. The lists of co-expressed genes were annotated using a unified re-annotation procedure. These gene lists were compared to find significant overlaps between studies. Conclusions Applied to this large compendium of data sets, meta-analyses demonstrated that conserved patterns between species could be identified. Focusing on a specific pathology (Duchenne Muscular Dystrophy) we validated results across independent studies and revealed robust biomarkers and new pathways of interest. The meta-analyses performed with MADMuscle show the usefulness of this approach. Our method can be applied to all public transcriptome data.
Collapse
|
165
|
CD8(+) T cells in facioscapulohumeral muscular dystrophy patients with inflammatory features at muscle MRI. J Clin Immunol 2010; 31:155-66. [PMID: 21063901 DOI: 10.1007/s10875-010-9474-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Accepted: 09/28/2010] [Indexed: 01/16/2023]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an inherited disease, and although strongly suggested, a contribution of inflammation to its pathogenesis has never been demonstrated. In FSHD patients, we found by immunohistochemistry inflammatory infiltrates mainly composed by CD8(+) T cells in muscles showing hyperintensity features on T2-weighted short tau inversion recovery magnetic resonance imaging (T2-STIR-MRI) sequences. Therefore, we evaluated the presence of circulating activated immune cells and the production of cytokines in patients with or without muscles showing hyperintensity features on T2-STIR-MRI sequences and from controls. FSHD patients displaying hyperintensity features in one or more muscles showed higher CD8(+)pSTAT1(+), CD8(+)T-bet(+) T cells and CD14(+)pSTAT1(+), CD14(+)T-bet(+) cells percentages and IL12p40, IFNγ and TNFα levels than patients without muscles displaying hyperintense features and controls. Moreover, the percentages of CD8(+)pSTAT1(+), CD8(+)T-bet(+) and CD14(+)pSTAT1(+) cells correlated with the proportion of muscles displaying hyperintensity features at T2-STIR sequences. These data indicate that circulating activated immune cells, mainly CD8(+) T cells, may favour FSHD progression by promoting active phases of muscle inflammation.
Collapse
|
166
|
Shi S, Hoogaars WMH, de Gorter DJJ, van Heiningen SH, Lin HY, Hong CC, Kemaladewi DU, Aartsma-Rus A, ten Dijke P, 't Hoen PAC. BMP antagonists enhance myogenic differentiation and ameliorate the dystrophic phenotype in a DMD mouse model. Neurobiol Dis 2010; 41:353-60. [PMID: 20940052 DOI: 10.1016/j.nbd.2010.10.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 09/23/2010] [Accepted: 10/01/2010] [Indexed: 12/29/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked lethal muscle wasting disease characterized by muscle fiber degeneration and necrosis. The progressive pathology of DMD can be explained by an insufficient regenerative response resulting in fibrosis and adipose tissue formation. BMPs are known to inhibit myogenic differentiation and in a previous study we found an increased expression of a BMP family member BMP4 in DMD myoblasts. The aim of the current study was therefore to investigate whether inhibition of BMP signaling could be beneficial for myoblast differentiation and muscle regeneration processes in a DMD context. All tested BMP inhibitors, Noggin, dorsomorphin and LDN-193189, were able to accelerate and enhance myogenic differentiation. However, dorsomorphin repressed both BMP and TGFβ signaling and was found to be toxic to primary myoblast cell cultures. In contrast, Noggin was found to be a potent and selective BMP inhibitor and was therefore tested in vivo in a DMD mouse model. Local adenoviral-mediated overexpression of Noggin in muscle resulted in an increased expression of the myogenic regulatory genes Myog and Myod1 and improved muscle histology. In conclusion, our results suggest that repression of BMP signaling may constitute an attractive adjunctive therapy for DMD patients.
Collapse
Affiliation(s)
- SongTing Shi
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Tee JM, Peppelenbosch MP. Anchoring skeletal muscle development and disease: the role of ankyrin repeat domain containing proteins in muscle physiology. Crit Rev Biochem Mol Biol 2010; 45:318-30. [PMID: 20515317 PMCID: PMC2942773 DOI: 10.3109/10409238.2010.488217] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The ankyrin repeat is a protein module with high affinity for other ankyrin repeats based on strong Van der Waals forces. The resulting dimerization is unusually resistant to both mechanical forces and alkanization, making this module exceedingly useful for meeting the extraordinary demands of muscle physiology. Many aspects of muscle function are controlled by the superfamily ankyrin repeat domain containing proteins, including structural fixation of the contractile apparatus to the muscle membrane by ankyrins, the archetypical member of the family. Additionally, other ankyrin repeat domain containing proteins critically control the various differentiation steps during muscle development, with Notch and developmental stage-specific expression of the members of the Ankyrin repeat and SOCS box (ASB) containing family of proteins controlling compartment size and guiding the various steps of muscle specification. Also, adaptive responses in fully formed muscle require ankyrin repeat containing proteins, with Myotrophin/V-1 ankyrin repeat containing proteins controlling the induction of hypertrophic responses following excessive mechanical load, and muscle ankyrin repeat proteins (MARPs) acting as protective mechanisms of last resort following extreme demands on muscle tissue. Knowledge on mechanisms governing the ordered expression of the various members of superfamily of ankyrin repeat domain containing proteins may prove exceedingly useful for developing novel rational therapy for cardiac disease and muscle dystrophies.
Collapse
Affiliation(s)
- Jin-Ming Tee
- Hubrecht Institute for Developmental Biology and Stem Cell Research-University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | | |
Collapse
|
168
|
Ghahramani Seno MM, Trollet C, Athanasopoulos T, Graham IR, Hu P, Dickson G. Transcriptomic analysis of dystrophin RNAi knockdown reveals a central role for dystrophin in muscle differentiation and contractile apparatus organization. BMC Genomics 2010; 11:345. [PMID: 20515474 PMCID: PMC2890566 DOI: 10.1186/1471-2164-11-345] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 06/01/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. DMD has a complex and as yet incompletely defined molecular pathophysiology hindering development of effective ameliorative approaches. Transcriptomic studies so far conducted on dystrophic cells and tissues suffer from non-specific changes and background noise due to heterogeneous comparisons and secondary pathologies. A study design in which a perfectly matched control cell population is used as reference for transcriptomic studies will give a much more specific insight into the effects of dystrophin deficiency and DMD pathophysiology. RESULTS Using RNA interference (RNAi) to knock down dystrophin in myotubes from C57BL10 mice, we created a homogenous model to study the transcriptome of dystrophin-deficient myotubes. We noted significant differences in the global gene expression pattern between these myotubes and their matched control cultures. In particular, categorical analyses of the dysregulated genes demonstrated significant enrichment of molecules associated with the components of muscle cell contractile unit, ion channels, metabolic pathways and kinases. Additionally, some of the dysregulated genes could potentially explain conditions and endophenotypes associated with dystrophin deficiency, such as dysregulation of calcium homeostasis (Pvalb and Casq1), or cardiomyopathy (Obscurin, Tcap). In addition to be validated by qPCR, our data gains another level of validity by affirmatively reproducing several independent studies conducted previously at genes and/or protein levels in vivo and in vitro. CONCLUSION Our results suggest that in striated muscles, dystrophin is involved in orchestrating proper development and organization of myofibers as contractile units, depicting a novel pathophysiology for DMD where the absence of dystrophin results in maldeveloped myofibers prone to physical stress and damage. Therefore, it becomes apparent that any gene therapy approaches for DMD should target early stages in muscle development to attain a maximum clinical benefit. With a clear and specific definition of the transcriptome of dystrophin deficiency, manipulation of identified dysregulated molecules downstream of dystrophin may lead to novel ameliorative approaches for DMD.
Collapse
|
169
|
Evans NP, Misyak SA, Robertson JL, Bassaganya-Riera J, Grange RW. Immune-mediated mechanisms potentially regulate the disease time-course of duchenne muscular dystrophy and provide targets for therapeutic intervention. PM R 2009; 1:755-68. [PMID: 19695529 DOI: 10.1016/j.pmrj.2009.04.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 04/23/2009] [Accepted: 04/28/2009] [Indexed: 11/19/2022]
Abstract
Duchenne muscular dystrophy is a lethal muscle-wasting disease that affects boys. Mutations in the dystrophin gene result in the absence of the dystrophin glycoprotein complex (DGC) from muscle plasma membranes. In healthy muscle fibers, the DGC forms a link between the extracellular matrix and the cytoskeleton to protect against contraction-induced membrane lesions and to regulate cell signaling. The absence of the DGC results in aberrant regulation of inflammatory signaling cascades. Inflammation is a key pathological characteristic of dystrophic muscle lesion formation. However, the role and regulation of this process in the disease time-course has not been sufficiently examined. The transcription factor nuclear factor-kappaB has been shown to contribute to the disease process and is likely involved with increased inflammatory gene expression, including cytokines and chemokines, found in dystrophic muscle. These aberrant signaling processes may regulate the early time-course of inflammatory events that contribute to the onset of disease. This review critically evaluates the possibility that dystrophic muscle lesions in both patients with Duchenne muscular dystrophy and mdx mice are the result of immune-mediated mechanisms that are regulated by inflammatory signaling and also highlights new therapeutic directions.
Collapse
Affiliation(s)
- Nicholas P Evans
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0002, USA.
| | | | | | | | | |
Collapse
|
170
|
Bhatnagar S, Kumar A. Therapeutic targeting of signaling pathways in muscular dystrophy. J Mol Med (Berl) 2009; 88:155-66. [PMID: 19816663 DOI: 10.1007/s00109-009-0550-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 09/28/2009] [Accepted: 09/29/2009] [Indexed: 02/06/2023]
Abstract
Muscular dystrophy refers to a group of genetic diseases that cause severe muscle weakness and loss of skeletal muscle mass. Although research has helped understanding the molecular basis of muscular dystrophy, there is still no cure for this devastating disorder. Numerous lines of investigation suggest that the primary deficiency of specific proteins causes aberrant activation of several cell signaling pathways in skeletal and cardiac muscle leading to the pathogenesis of muscular dystrophy. Studies using genetic mouse models and pharmacological approaches have provided strong evidence that the modulation of the activity of specific cell signaling pathways has enormous potential to improving the quality of life and extending the life expectancy in muscular dystrophy patients. In this article, we have outlined the current understanding regarding the role of different cell signaling pathways in disease progression with particular reference to different models of muscular dystrophy and the development of therapy.
Collapse
Affiliation(s)
- Shephali Bhatnagar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, 500 South Preston Street, Louisville, KY, 40202, USA
| | | |
Collapse
|
171
|
Colussi C, Gurtner A, Rosati J, Illi B, Ragone G, Piaggio G, Moggio M, Lamperti C, D'Angelo G, Clementi E, Minetti G, Mozzetta C, Antonini A, Capogrossi MC, Puri PL, Gaetano C. Nitric oxide deficiency determines global chromatin changes in Duchenne muscular dystrophy. FASEB J 2009; 23:2131-41. [PMID: 19264835 DOI: 10.1096/fj.08-115618] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The present study provides evidence that abnormal patterns of global histone modification are present in the skeletal muscle nuclei of mdx mice and Duchenne muscular dystrophy (DMD) patients. A combination of specific histone H3 modifications, including Ser-10 phosphorylation, acetylation of Lys 9 and 14, and Lys 79 methylation, were found enriched in muscle biopsies from human patients affected by DMD and in late-term fetuses, early postnatal pups, or adult mdx mice. In this context, chromatin immunoprecipitation experiments showed an enrichment of these modifications at the loci of genes involved in proliferation or inflammation, suggesting a regulatory effect on gene expression. Remarkably, the reexpression of dystrophin induced by gentamicin treatment or the administration of nitric oxide (NO) donors reversed the abnormal pattern of H3 histone modifications. These findings suggest an unanticipated link between the dystrophin-activated NO signaling and the remodeling of chromatin. In this context, the regulation of class IIa histone deacetylases (HDACs) 4 and 5 was found altered as a consequence of the reduced NO-dependent protein phosphatase 2A activity, indicating that both NO and class IIa HDACs are important for satellite cell differentiation and gene expression in mdx mice. In conclusion, this work provides the first evidence of a role for NO as an epigenetic regulator in DMD.
Collapse
Affiliation(s)
- Claudia Colussi
- Laboratorio di Terapia Genica e Biologia Vascolare, Istituto Cardiologico Monzino, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Andersen DC, Petersson SJ, Jørgensen LH, Bollen P, Jensen PB, Teisner B, Schroeder HD, Jensen CH. Characterization of DLK1+ cells emerging during skeletal muscle remodeling in response to myositis, myopathies, and acute injury. Stem Cells 2009; 27:898-908. [PMID: 19353518 DOI: 10.1634/stemcells.2008-0826] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Delta like 1 (DLK1) has been proposed to act as a regulator of cell fate determination and is linked to the development of various tissues including skeletal muscle. Herein we further investigated DLK1 expression during skeletal muscle remodeling. Although practically absent in normal adult muscle, DLK1 was upregulated in all human myopathies analyzed, including Duchenne- and Becker muscular dystrophies. Substantial numbers of DLK1(+) satellite cells were observed in normal neonatal and Duchenne muscle, and furthermore, myogenic DLK1(+) cells were identified during muscle regeneration in animal models in which the peak expression of Dlk1 mRNA and protein coincided with that of myoblast differentiation and fusion. In addition to perivascular DLK1(+) cells, interstitial DLK1(+) cells were numerous in regenerating muscle, and in agreement with colocalization studies of DLK1 and CD90/DDR2, qPCR of fluorescence-activated cell sorting DLK1(+) and DLK1(-) cells revealed that the majority of DLK1(+) cells isolated at day 7 of regeneration had a fibroblast-like phenotype. The existence of different DLK1(+) populations was confirmed in cultures of primary derived myogenic cells, in which large flat nonmyogenic DLK1(+) cells and small spindle-shaped cells coexpressing DLK1 and muscle-specific markers were observed. Myogenic differentiation was achieved when sorted DLK1(+) cells were cocultured together with primary myoblasts revealing a myogenic potential that was 10% of the DLK1(-) population. Transplantation of DLK1(+) cells into lacerated muscle did, however, not give rise to DLK1(+) cell-derived myofibers. We suggest that the DLK1(+) subpopulations identified herein each may contribute at different levels/time points to the processes involved in muscle development and remodeling.
Collapse
Affiliation(s)
- Ditte C Andersen
- Department of Clinical Pathology, Odense University Hospital, Odense C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
173
|
Greco S, De Simone M, Colussi C, Zaccagnini G, Fasanaro P, Pescatori M, Cardani R, Perbellini R, Isaia E, Sale P, Meola G, Capogrossi MC, Gaetano C, Martelli F. Common micro‐RNA signature in skeletal muscle damage and regeneration induced by Duchenne muscular dystrophy and acute ischemia. FASEB J 2009; 23:3335-46. [DOI: 10.1096/fj.08-128579] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Simona Greco
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato San Donato Milanese Milan Italy
| | | | | | - Germana Zaccagnini
- Istituto Dermopatico dell'Immacolata—IRCCS and San Raffaele Pisana—IRCCS Rome Italy
| | - Pasquale Fasanaro
- Istituto Dermopatico dell'Immacolata—IRCCS and San Raffaele Pisana—IRCCS Rome Italy
| | | | | | - Riccardo Perbellini
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato San Donato Milanese Milan Italy
- Università di Milano Milan Italy
| | - Eleonora Isaia
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato San Donato Milanese Milan Italy
| | | | - Giovanni Meola
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato San Donato Milanese Milan Italy
- Università di Milano Milan Italy
| | | | - Carlo Gaetano
- Istituto Dermopatico dell'Immacolata—IRCCS and San Raffaele Pisana—IRCCS Rome Italy
| | - Fabio Martelli
- Istituto Dermopatico dell'Immacolata—IRCCS and San Raffaele Pisana—IRCCS Rome Italy
| |
Collapse
|
174
|
Dysregulated intracellular signaling and inflammatory gene expression during initial disease onset in Duchenne muscular dystrophy. Am J Phys Med Rehabil 2009; 88:502-22. [PMID: 19454857 DOI: 10.1097/phm.0b013e3181a5a24f] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Duchenne muscular dystrophy is a debilitating genetic disorder characterized by severe muscle wasting and early death in affected boys. The primary cause of this disease is mutations in the dystrophin gene that result in the absence of the protein dystrophin and the associated dystrophin-glycoprotein complex in the plasma membrane of muscle fibers. In normal muscle, this complex forms a link between the extracellular matrix and the cytoskeleton that is thought to protect muscle fibers from contraction-induced membrane lesions and to regulate cell signaling cascades. Although the primary defect is known, the mechanisms that initiate disease onset have not been characterized. Data collected during early maturation suggest that inflammatory and immune responses are key contributors to disease pathogenesis and may be initiated by aberrant signaling in dystrophic muscle. However, detailed time course studies of the inflammatory and immune processes are incomplete and need to be characterized further to understand the disease progression. The purposes of this review are to examine the possibility that initial disease onset in dystrophin-deficient muscle results from aberrant inflammatory signaling pathways and to highlight the potential clinical relevance of targeting these pathways to treat Duchenne muscular dystrophy.
Collapse
|
175
|
Zhang M, Zhang L, Zou J, Yao C, Xiao H, Liu Q, Wang J, Wang D, Wang C, Guo Z. Evaluating reproducibility of differential expression discoveries in microarray studies by considering correlated molecular changes. ACTA ACUST UNITED AC 2009; 25:1662-8. [PMID: 19417058 PMCID: PMC2940240 DOI: 10.1093/bioinformatics/btp295] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
MOTIVATION According to current consistency metrics such as percentage of overlapping genes (POG), lists of differentially expressed genes (DEGs) detected from different microarray studies for a complex disease are often highly inconsistent. This irreproducibility problem also exists in other high-throughput post-genomic areas such as proteomics and metabolism. A complex disease is often characterized with many coordinated molecular changes, which should be considered when evaluating the reproducibility of discovery lists from different studies. RESULTS We proposed metrics percentage of overlapping genes-related (POGR) and normalized POGR (nPOGR) to evaluate the consistency between two DEG lists for a complex disease, considering correlated molecular changes rather than only counting gene overlaps between the lists. Based on microarray datasets of three diseases, we showed that though the POG scores for DEG lists from different studies for each disease are extremely low, the POGR and nPOGR scores can be rather high, suggesting that the apparently inconsistent DEG lists may be highly reproducible in the sense that they are actually significantly correlated. Observing different discovery results for a disease by the POGR and nPOGR scores will obviously reduce the uncertainty of the microarray studies. The proposed metrics could also be applicable in many other high-throughput post-genomic areas.
Collapse
Affiliation(s)
- Min Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Colussi C, Mozzetta C, Gurtner A, Illi B, Rosati J, Straino S, Ragone G, Pescatori M, Zaccagnini G, Antonini A, Minetti G, Martelli F, Piaggio G, Gallinari P, Steinkuhler C, Clementi E, Dell'Aversana C, Altucci L, Mai A, Capogrossi MC, Puri PL, Gaetano C. HDAC2 blockade by nitric oxide and histone deacetylase inhibitors reveals a common target in Duchenne muscular dystrophy treatment. Proc Natl Acad Sci U S A 2008; 105:19183-7. [PMID: 19047631 PMCID: PMC2614736 DOI: 10.1073/pnas.0805514105] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Indexed: 12/25/2022] Open
Abstract
The overlapping histological and biochemical features underlying the beneficial effect of deacetylase inhibitors and NO donors in dystrophic muscles suggest an unanticipated molecular link among dystrophin, NO signaling, and the histone deacetylases (HDACs). Higher global deacetylase activity and selective increased expression of the class I histone deacetylase HDAC2 were detected in muscles of dystrophin-deficient MDX mice. In vitro and in vivo siRNA-mediated down-regulation of HDAC2 in dystrophic muscles was sufficient to replicate the morphological and functional benefits observed with deacetylase inhibitors and NO donors. We found that restoration of NO signaling in vivo, by adenoviral-mediated expression of a constitutively active endothelial NOS mutant in MDX muscles, and in vitro, by exposing MDX-derived satellite cells to NO donors, resulted in HDAC2 blockade by cysteine S-nitrosylation. These data reveal a special contribution of HDAC2 in the pathogenesis of Duchenne muscular dystrophy and indicate that HDAC2 inhibition by NO-dependent S-nitrosylation is important for the therapeutic response to NO donors in MDX mice. They also define a common target for independent pharmacological interventions in the treatment of Duchenne muscular dystrophy.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Cells, Cultured
- Enzyme Inhibitors/pharmacology
- Epigenesis, Genetic
- Histone Deacetylase 2
- Histone Deacetylase Inhibitors
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle, Skeletal/cytology
- Muscular Dystrophy, Animal/drug therapy
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts/cytology
- Myoblasts/enzymology
- Nitric Oxide/metabolism
- Nitrogen/metabolism
- Pyridines/pharmacology
- RNA, Small Interfering
- Repressor Proteins/antagonists & inhibitors
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/enzymology
Collapse
Affiliation(s)
- Claudia Colussi
- Laboratorio di Terapia Genica e Biologia Vascolare, Istituto Cardiologico Monzino, 20138 Milan, Italy
| | - Chiara Mozzetta
- Dulbecco Telethon Institute, European Brain Research Institute/Santa Lucia Foundation, 00143 Rome, Italy
| | - Aymone Gurtner
- Laboratorio di Oncogenesi Molecolare and Rome Oncogenomic Center, Istituto Regina Elena, 00158 Rome, Italy
| | - Barbara Illi
- Laboratorio di Terapia Genica e Biologia Vascolare, Istituto Cardiologico Monzino, 20138 Milan, Italy
| | | | | | - Gianluca Ragone
- Oncologia Molecolare, Istituto Dermopatico dell' Immacolata, 00167 Rome, Italy
| | | | | | | | - Giulia Minetti
- Dulbecco Telethon Institute, European Brain Research Institute/Santa Lucia Foundation, 00143 Rome, Italy
| | | | - Giulia Piaggio
- Laboratorio di Oncogenesi Molecolare and Rome Oncogenomic Center, Istituto Regina Elena, 00158 Rome, Italy
| | | | | | - Emilio Clementi
- Istituto Scientifico E. Medea, 23842 Bosisio Parini, Italy
- Dipartimento di Scienze Precliniche, Università di Milano, 20122 Milan, Italy
| | | | - Lucia Altucci
- Dipartimento di Patologia Generale, Seconda Università di Napoli, 80138 Napoli, Italy
| | - Antonello Mai
- Istituto Pasteur, Fondazione Cenci Bolognetti, Dipartimento di Studi Farmaceutici, Sapienza Università di Roma, 00185 Rome, Italy; and
| | | | - Pier Lorenzo Puri
- Dulbecco Telethon Institute, European Brain Research Institute/Santa Lucia Foundation, 00143 Rome, Italy
- The Burnham Institute for Medical Research, San Diego, CA 92037
| | | |
Collapse
|
177
|
Plotnikov SV, Kenny AM, Walsh SJ, Zubrowski B, Joseph C, Scranton VL, Kuchel GA, Dauser D, Xu M, Pilbeam CC, Adams DJ, Dougherty RP, Campagnola PJ, Mohler WA. Measurement of muscle disease by quantitative second-harmonic generation imaging. JOURNAL OF BIOMEDICAL OPTICS 2008; 13:044018. [PMID: 19021346 DOI: 10.1117/1.2967536] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Determining the health of muscle cells by in vivo imaging could impact the diagnosis and monitoring of a large number of congenital and acquired muscular or cardiac disorders. However, currently used technologies are hampered by insufficient resolution, lack of specificity, or invasiveness. We have combined intrinsic optical second-harmonic generation from sarcomeric myosin with a novel mathematical treatment of striation pattern analysis, to obtain measures of muscle contractile integrity that correlate strongly with the neuromuscular health of mice suffering from genetic, acquired, and age-related decline in skeletal muscle function. Analysis of biopsies from a pilot group of human volunteers suggests a similar power in quantifying sarcopenic changes in muscle integrity. These results provide the first strong evidence that quantitative image analysis of sarcomere pattern can be correlated with physiological function, and they invite the application of SHG imaging in clinical practice, either in biopsy samples or via microendoscopy.
Collapse
Affiliation(s)
- Sergey V Plotnikov
- University of Connecticut Health Center, Department of Genetics and Developmental Biology, Farmington, Connecticut 06030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Kadota K, Nakai Y, Shimizu K. A weighted average difference method for detecting differentially expressed genes from microarray data. Algorithms Mol Biol 2008; 3:8. [PMID: 18578891 PMCID: PMC2464587 DOI: 10.1186/1748-7188-3-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Accepted: 06/26/2008] [Indexed: 01/05/2023] Open
Abstract
Background Identification of differentially expressed genes (DEGs) under different experimental conditions is an important task in many microarray studies. However, choosing which method to use for a particular application is problematic because its performance depends on the evaluation metric, the dataset, and so on. In addition, when using the Affymetrix GeneChip® system, researchers must select a preprocessing algorithm from a number of competing algorithms such as MAS, RMA, and DFW, for obtaining expression-level measurements. To achieve optimal performance for detecting DEGs, a suitable combination of gene selection method and preprocessing algorithm needs to be selected for a given probe-level dataset. Results We introduce a new fold-change (FC)-based method, the weighted average difference method (WAD), for ranking DEGs. It uses the average difference and relative average signal intensity so that highly expressed genes are highly ranked on the average for the different conditions. The idea is based on our observation that known or potential marker genes (or proteins) tend to have high expression levels. We compared WAD with seven other methods; average difference (AD), FC, rank products (RP), moderated t statistic (modT), significance analysis of microarrays (samT), shrinkage t statistic (shrinkT), and intensity-based moderated t statistic (ibmT). The evaluation was performed using a total of 38 different binary (two-class) probe-level datasets: two artificial "spike-in" datasets and 36 real experimental datasets. The results indicate that WAD outperforms the other methods when sensitivity and specificity are considered simultaneously: the area under the receiver operating characteristic curve for WAD was the highest on average for the 38 datasets. The gene ranking for WAD was also the most consistent when subsets of top-ranked genes produced from three different preprocessed data (MAS, RMA, and DFW) were compared. Overall, WAD performed the best for MAS-preprocessed data and the FC-based methods (AD, WAD, FC, or RP) performed well for RMA and DFW-preprocessed data. Conclusion WAD is a promising alternative to existing methods for ranking DEGs with two classes. Its high performance should increase researchers' confidence in microarray analyses.
Collapse
|
179
|
TCAP knockdown by RNA interference inhibits myoblast differentiation in cultured skeletal muscle cells. Neuromuscul Disord 2008; 18:413-22. [PMID: 18440815 DOI: 10.1016/j.nmd.2008.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 02/29/2008] [Accepted: 03/11/2008] [Indexed: 11/20/2022]
Abstract
Null mutation of titin-cap (TCAP) causes limb-girdle muscular dystrophy type 2G (LGMD2G). LGMD2G patients develop muscle atrophy, and lose the ability to walk by their third decade. Previous findings suggest that TCAP regulates myostatin, a key regulator of muscle growth. We tested the hypothesis that TCAP knockdown with RNA interference will lead to differential expression of genes involved in muscle proliferation and differentiation, impairing muscle cell growth. mRNA from cultured cells treated with TCAP siRNA duplex constructs was analyzed using Northern blots and real-time RT-PCR. siRNA treatment decreased TCAP mRNA expression in differentiating muscle cells. Significant (p<0.05) decreases in mRNA were observed for myogenic regulatory factors. siRNA treatment also prevented development of the normal phenotype of muscle cells. Our findings suggest that TCAP knockdown with RNA interference alters normal muscle cell differentiation.
Collapse
|
180
|
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy of childhood. This incurable disease is characterised by muscle wasting and loss of walking ability leading to complete wheelchair dependence by 13 years of age. Prolongation of walking is one of the major aims of treatment. OBJECTIVES The aim of this review was to assess whether glucocorticoid corticosteroids stabilize or improve muscle strength and walking in boys with DMD. SEARCH STRATEGY This is an update of the Cochrane systematic review first published in 2004 (Manzur 2004). We searched the Cochrane Neuromuscular Disease Group Trials Register (August 2006) using the term 'Duchenne muscular dystrophy'. We also searched MEDLINE (January 1966 to July 2007), EMBASE (January 1980 to August 2006), CINAHL and LILACS (January 1982 to August 2006). We wrote to authors of published studies and other experts in this disease to help identify other trials, checked the references in the identified trials and hand searched the abstracts of relevant journals. SELECTION CRITERIA Types of studies: randomised or quasi-randomised trials. TYPES OF PARTICIPANTS all patients with a definite diagnosis of Duchenne muscular dystrophy. Types of interventions: glucocorticoids such as prednisone, prednisolone, deflazacort or others, with a minimum treatment period of three months. PRIMARY OUTCOME MEASURE prolongation of walking (independent walking without long leg calipers). SECONDARY OUTCOME MEASURES strength outcome measures, manual muscle strength testing using Medical Research Council strength scores, functional outcome measures and adverse events. DATA COLLECTION AND ANALYSIS We identified six randomised controlled trials that met the inclusion criteria for our review, and one of these (Beenakker 2005) is a new addition to this update, as it was published subsequent to our first review (Manzur 2004). Two review authors independently selected the trials for the review and assessed methodological quality. Data extraction and inputting were double-checked. PRIMARY OUTCOME MEASURE data from one small study used prolongation of walking as an outcome measure and did not show significant benefit. SECONDARY OUTCOME MEASURES The meta-analysis of the results from four randomised controlled trials with altogether 249 participants showed that glucocorticoid corticosteroids improved muscle strength and function over six months. Improvements were seen in time taken to rise from the floor (Gowers' time), nine metres walking time, four-stair climbing time, ability to lift weights, leg function grade and forced vital capacity. One randomised controlled trial with altogether 28 participants showed that glucocorticoid corticosteroids stabilize muscle strength and function for up to two years. The most effective prednisolone regime appears to be 0.75 mg/kg/day, given in a daily dose regime. Not enough data were available to compare efficacy of prednisone with deflazacort. Adverse effects: Excessive weight gain, behavioural abnormalities, cushingoid appearance and excessive hair growth were all more common with glucocorticoid corticosteroids than placebo. Long-term adverse effects of glucocorticoid therapy could not be evaluated because of the short-term duration of the randomised studies.Non-randomised studies: A number of non-randomised studies with important efficacy and adverse effects data are tabulated and discussed. AUTHORS' CONCLUSIONS There is evidence from randomised controlled studies that glucocorticoid corticosteroid therapy in Duchenne muscular dystrophy improves muscle strength and function in the short-term (six months to two years). The most effective prednisolone regime appears to be 0.75 mg/kg/day, given daily. In the short term, adverse effects were significantly more common but not clinically severe. Long-term benefits and hazards of glucocorticoid treatment cannot be evaluated from the currently published randomised studies. Non-randomised studies support the conclusions of functional benefits but also identify clinically significant adverse effects of long-term treatment. These benefits and adverse effects have implications for future research studies and clinical practice.
Collapse
Affiliation(s)
- A Y Manzur
- Hammersmith Hospital, Dubowitz Neuromuscular Centre, Department of Paediatrics, DuCane Road, London, UK, W12 OHS.
| | | | | | | |
Collapse
|
181
|
Radley HG, Davies MJ, Grounds MD. Reduced muscle necrosis and long-term benefits in dystrophic mdx mice after cV1q (blockade of TNF) treatment. Neuromuscul Disord 2008; 18:227-38. [PMID: 18207402 DOI: 10.1016/j.nmd.2007.11.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 10/10/2007] [Accepted: 11/01/2007] [Indexed: 11/26/2022]
Abstract
Tumour necrosis factor (TNF) is a potent inflammatory cytokine that appears to exacerbate damage of dystrophic muscle in vivo. The monoclonal murine specific antibody cV1q that specifically neutralises murine TNF demonstrated significant anti-inflammatory effects in dystrophic mdx mice. cV1q administration protected dystrophic skeletal myofibres against necrosis in both young and adult mdx mice and in adult mdx mice subjected to 48 h voluntary wheel exercise. Long-term studies (up to 90 days) in voluntarily exercised mdx mice showed beneficial effects of cV1q treatment with reduced histological evidence of myofibre damage and a striking decrease in serum creatine kinase levels. However, in the absence of exercise long-term cV1q treatment did not reduce necrosis or background pathology in mdx mice. An additional measure of well-being in the cV1q treated mice was that they ran significantly more than control mdx mice.
Collapse
Affiliation(s)
- Hannah G Radley
- School of Anatomy and Human Biology, The University of Western Australia, Anatomy and Human Biology, Perth, WA 6009, Australia
| | | | | |
Collapse
|
182
|
Abstract
PURPOSE OF REVIEW To illustrate potential risks and benefits of disease screening for newborns using the example of Duchenne muscular dystrophy. RECENT FINDINGS There is a wide range in the reported positive predictive value for screening male newborns for Duchenne muscular dystrophy by the creatine kinase level on dried blood spots. Some parental anxiety is associated with both early detection and false-positive screening results. No data are available about the impact of the diagnosis on the child, including the impact of early initiation of therapy. Studies suggest that few parents change their future reproductive planning based on identification of Duchenne muscular dystrophy through screening. Few data are available regarding the cost of newborn screening for Duchenne muscular dystrophy, and there are insufficient data to evaluate the cost-effectiveness of Duchenne muscular dystrophy screening. SUMMARY Available data are insufficient to recommend routine newborn screening for Duchenne muscular dystrophy. Understanding the gaps in knowledge provides insight into the evidence needed to recommend newborn screening for Duchenne muscular dystrophy. Studies are needed to evaluate the potential risks and benefits of screening, including the associated incremental costs.
Collapse
|
183
|
Zanotti S, Saredi S, Ruggieri A, Fabbri M, Blasevich F, Romaggi S, Morandi L, Mora M. Altered extracellular matrix transcript expression and protein modulation in primary Duchenne muscular dystrophy myotubes. Matrix Biol 2007; 26:615-24. [PMID: 17662584 DOI: 10.1016/j.matbio.2007.06.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 06/11/2007] [Accepted: 06/20/2007] [Indexed: 11/19/2022]
Abstract
Extent of muscle fibrosis contributes to disease severity in muscular dystrophies. To investigate whether extracellular matrix (ECM) components contribute to the severe fibrosis observed in Duchenne muscular dystrophy (DMD) skeletal muscle, we quantitated several ECM components (transcripts and proteins) in primary DMD and control myotube cultures. We evaluated the fibrogenic transforming growth factor- beta1 (TGF-beta1); the small pleiotropic proteoglycan decorin, involved in collagen fibrillogenesis and TGF-beta1 modulation; metalloproteinases MMP-2 and MMP-9; tissue inhibitors of metalloproteinase (TIMP) 1, 2 and 3; collagens I and VI; and the tissue factor myostatin that inhibits muscle growth. Dystrophic myotube cultures had significantly lower levels of decorin mRNA, as also observed in DMD muscle biopsies, and significantly higher levels of TGF-beta1, myostatin, and collagens I and VI. MMP-2, TIMP-1 and TIMP-2 transcript levels were also significantly increased in DMD, but MMP-9 and TIMP-3 transcripts were unchanged. By zymography, MMP-2 activity was significantly higher in DMD than control. Protein levels were similar in DMD and controls but myostatin protein was significantly increased in DMD. We have found that transcript expression and protein modulation of several ECM components is altered in DMD muscle cells in vitro, indicating that these cells contribute fundamentally to the pathological process, since the inflammation and degeneration characterizing DMD muscle in vivo are presumably absent in culture. Our findings that myostatin-potent inhibitor of satellite cell activation and muscle renewal--is increased, and that decorin-binder and downregulator of TGFbeta1 and myostatin--is decreased, may have implications for DMD therapy to reduce muscle fibrosis.
Collapse
Affiliation(s)
- S Zanotti
- Neuromuscular Diseases and Neuroimmunology Unit, Foundation IRCCS Neurological Institute C. Besta, Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|