101
|
de Senzi Moraes Pinto R, Ferretti R, Moraes LHR, Neto HS, Marques MJ, Minatel E. N-acetylcysteine treatment reduces TNF-α levels and myonecrosis in diaphragm muscle of mdx mice. Clin Nutr 2012; 32:472-5. [PMID: 22727548 DOI: 10.1016/j.clnu.2012.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 05/08/2012] [Accepted: 06/01/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND & AIMS Duchenne muscular dystrophy (DMD) is a genetic muscle disease caused by the absence of dystrophin. An established animal model of DMD is the mdx mouse, which is unable to express dystrophin. Inflammation, particularly the proinflammatory cytokine tumor necrosis factor alpha (TNF-α), strongly contributes to necrosis in the dystrophin-deficient fibers of the mdx mice and in DMD. In this study we investigated whether the antioxidant N-acetylcysteine (NAC) decreases TNF-α levels and protects the diaphragm muscle of mdx mice against necrosis. METHODS Mdx mice (14 days old) received daily intraperitoneal injections of NAC for 14 days, followed by removal of the diaphragm muscle. Control mdx mice were injected with saline. RESULTS NAC reduced TNF-α and 4-HNE-protein adducts levels, inflammation, creatine kinase levels, and myonecrosis in diaphragm muscle. CONCLUSIONS NAC may be used as a complementary treatment for dystrophinopathies. However, clinical trials are needed to determine the appropriate dose for patients with Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Rafael de Senzi Moraes Pinto
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | | | | | | | | | | |
Collapse
|
102
|
Tuazon MA, Henderson GC. Fatty acid profile of skeletal muscle phospholipid is altered in mdx mice and is predictive of disease markers. Metabolism 2012; 61:801-11. [PMID: 22209669 DOI: 10.1016/j.metabol.2011.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 10/10/2011] [Accepted: 10/21/2011] [Indexed: 10/14/2022]
Abstract
The mdx mouse is a model for Duchenne muscular dystrophy. The fatty acid (FA) composition in dystrophic muscle could potentially impact the disease severity. We tested FA profiles in skeletal muscle phospholipid (PL) and triglyceride in mdx and control (con) mice to assess associations with disease state as well as correlations with grip strength (which is lower in mdx) and serum creatine kinase (CK, which is elevated in mdx). Compared with con, mdx PL contained less docosahexaenoic acid (P < .001) and more linoleic acid (P = .001). Docosahexaenoic acid contents did not correlate with strength or serum CK. Linoleic acid content in PL was positively correlated with CK in mdx (P < .05) but not con. α-Linolenic acid content in PL was positively correlated with strength in mdx (P < .05) but not con. The FA profile in triglyceride showed less difference between groups and far less predictive ability for disease markers. We conclude that profiling the FA composition of tissue lipids (particularly PL) can be a useful strategy for generating novel biomarkers and potential therapeutic targets in muscle diseases and likely other pathological conditions as well. Specifically, the present results have indicated potential benefits of raising content of particular n-3 FAs (especially α-linolenic acid) and reducing content of particular n-6 FAs (linoleic acid) in PL of dystrophic muscle.
Collapse
Affiliation(s)
- Marc A Tuazon
- Department of Exercise Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08901, USA
| | | |
Collapse
|
103
|
Tonon E, Ferretti R, Shiratori JH, Santo Neto H, Marques MJ, Minatel E. Ascorbic acid protects the diaphragm muscle against myonecrosis in mdx mice. Nutrition 2012; 28:686-90. [DOI: 10.1016/j.nut.2011.09.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 08/09/2011] [Accepted: 09/12/2011] [Indexed: 11/30/2022]
|
104
|
Vidal B, Ardite E, Suelves M, Ruiz-Bonilla V, Janué A, Flick MJ, Degen JL, Serrano AL, Muñoz-Cánoves P. Amelioration of Duchenne muscular dystrophy in mdx mice by elimination of matrix-associated fibrin-driven inflammation coupled to the αMβ2 leukocyte integrin receptor. Hum Mol Genet 2012; 21:1989-2004. [PMID: 22381526 DOI: 10.1093/hmg/dds012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), a persistently altered and reorganizing extracellular matrix (ECM) within inflamed muscle promotes damage and dysfunction. However, the molecular determinants of the ECM that mediate inflammatory changes and faulty tissue reorganization remain poorly defined. Here, we show that fibrin deposition is a conspicuous consequence of muscle-vascular damage in dystrophic muscles of DMD patients and mdx mice and that elimination of fibrin(ogen) attenuated dystrophy progression in mdx mice. These benefits appear to be tied to: (i) a decrease in leukocyte integrin α(M)β(2)-mediated proinflammatory programs, thereby attenuating counterproductive inflammation and muscle degeneration; and (ii) a release of satellite cells from persistent inhibitory signals, thereby promoting regeneration. Remarkably, Fib-gamma(390-396A) (Fibγ(390-396A)) mice expressing a mutant form of fibrinogen with normal clotting function, but lacking the α(M)β(2) binding motif, ameliorated dystrophic pathology. Delivery of a fibrinogen/α(M)β(2) blocking peptide was similarly beneficial. Conversely, intramuscular fibrinogen delivery sufficed to induce inflammation and degeneration in fibrinogen-null mice. Thus, local fibrin(ogen) deposition drives dystrophic muscle inflammation and dysfunction, and disruption of fibrin(ogen)-α(M)β(2) interactions may provide a novel strategy for DMD treatment.
Collapse
Affiliation(s)
- Berta Vidal
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University, CIBER on Neurodegenerative Diseases, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Nakagaki WR, Camilli JA. Spontaneous Healing Capacity of Calvarial Bone Defects in mdx Mice. Anat Rec (Hoboken) 2012; 295:590-6. [DOI: 10.1002/ar.22412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/20/2011] [Indexed: 11/09/2022]
|
106
|
Nakagaki WR, Camilli JA. Bone tissue and muscle dystrophin deficiency in mdx mice. Joint Bone Spine 2011; 79:129-33. [PMID: 22079415 DOI: 10.1016/j.jbspin.2011.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 08/08/2011] [Indexed: 01/06/2023]
Abstract
Duchenne muscular dystrophy is a neuromuscular disease caused by the lack of dystrophin that affects skeletal muscles, causing degeneration of muscle fibers and replacing them with fibrous and adipose tissue, events that gradually lead to functional loss. Patients with Duchenne muscular dystrophy have shown that bones become more fragile with age and with advancement of the disease. Muscle weakness and reduced mobility have been suggested to be the factors that promote bone deterioration. However, it seems that this does not occur in mdx mice. It has been identified in mdx mice the existence of a factor related or not to the lack of dystrophin that also participates in the impairment of bone quality. Mdx mice also exhibit muscle degeneration, but unlike human, it is compensated by muscle regeneration. In consequence, there is an increase in the muscle mass, but not necessarily of muscle contractile strength. The accommodation of this increased muscle mass promotes bone formation at specific sites, such as at tendo-osseous junctions. In addition, the inflammatory response to muscle injury may be responsible for the increase in angiogenesis and regeneration observed in mdx mice, inducing the release of cytokines and chemokines that play an important role in the recruitment of leukocytes and macrophages. Then, mdx mice may possess compensatory mechanisms in bone in response to a genetic defect.
Collapse
Affiliation(s)
- Wilson Romero Nakagaki
- Department of Anatomy, Cell Biology and Physiology and Biophysics, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | |
Collapse
|
107
|
Charan RA, Hanson R, Clemens PR. Deubiquitinating enzyme A20 negatively regulates NF-κB signaling in skeletal muscle in mdx mice. FASEB J 2011; 26:587-95. [PMID: 22012122 DOI: 10.1096/fj.11-189829] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by the lack of a functional dystrophin protein that results in muscle fiber membrane disruption and, ultimately, degeneration. Regeneration of muscle fibers fails progressively, and muscle tissue is replaced with connective tissue. As a result, DMD causes progressive limb muscle weakness and cardiac and respiratory failure. The absence of dystrophin from muscle fibers triggers the chronic activation of the nuclear factor of kappa B (NF-κB). Chronic activation of NF-κB in muscle leads to infiltration of macrophages, up-regulation of the ubiquitin-proteosome system, and down-regulation of the helix-loop-helix muscle regulatory factor, MyoD. These processes, triggered by NF-κB activation, promote muscle degeneration and failure of muscle regeneration. A20 (TNFAIP3) is a critical negative regulator of NF-κB. In this study, we characterize the role of A20 in regulating NF-κB activation in skeletal muscle, identifying a novel role in muscle regeneration. A20 is highly expressed in regenerating muscle fibers, and knockdown of A20 impairs muscle differentiation in vitro, which suggests that A20 expression is critically important for regeneration of dystrophic muscle tissue. Furthermore, down-regulation of the classic pathway of NF-κB activation is associated with up-regulation of the alternate pathway in regenerating muscle fibers, suggesting a mechanism by which A20 promotes muscle regeneration. These results demonstrate the important role of A20 in muscle fiber repair and suggest the potential of A20 as a therapeutic target to ameliorate the pathology and clinical symptoms of DMD.
Collapse
Affiliation(s)
- Rakshita A Charan
- Neurology Service, Department of Veterans Affairs Medical Center, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
108
|
|
109
|
Reay DP, Yang M, Watchko JF, Daood M, O'Day TL, Rehman KK, Guttridge DC, Robbins PD, Clemens PR. Systemic delivery of NEMO binding domain/IKKγ inhibitory peptide to young mdx mice improves dystrophic skeletal muscle histopathology. Neurobiol Dis 2011; 43:598-608. [PMID: 21624467 PMCID: PMC3145633 DOI: 10.1016/j.nbd.2011.05.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 04/01/2011] [Accepted: 05/14/2011] [Indexed: 10/18/2022] Open
Abstract
The activation of nuclear factor κB (NF-κB) contributes to muscle degeneration that results from dystrophin deficiency in human Duchenne muscular dystrophy (DMD) and in the mdx mouse. In dystrophic muscle, NF-κB participates in inflammation and failure of muscle regeneration. Peptides containing the NF-κB Essential Modulator (NEMO) binding domain (NBD) disrupt the IκB kinase complex, thus blocking NF-κB activation. The NBD peptide, which is linked to a protein transduction domain to achieve in vivo peptide delivery to muscle tissue, was systemically delivered to mdx mice for 4 or 7 weeks to study NF-κB activation, histological changes in hind limb and diaphragm muscle and ex vivo function of diaphragm muscle. Decreased NF-κB activation, decreased necrosis and increased regeneration were observed in hind limb and diaphragm muscle in mdx mice treated systemically with NBD peptide, as compared to control mdx mice. NBD peptide treatment resulted in improved generation of specific force and greater resistance to lengthening activations in diaphragm muscle ex vivo. Together these data support the potential of NBD peptides for the treatment of DMD by modulating dystrophic pathways in muscle that are downstream of dystrophin deficiency.
Collapse
MESH Headings
- Animals
- Diaphragm/pathology
- Diaphragm/physiology
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- I-kappa B Kinase/administration & dosage
- I-kappa B Kinase/pharmacokinetics
- I-kappa B Kinase/therapeutic use
- Intracellular Signaling Peptides and Proteins/administration & dosage
- Intracellular Signaling Peptides and Proteins/pharmacokinetics
- Intracellular Signaling Peptides and Proteins/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle Contraction/drug effects
- Muscle Contraction/physiology
- Muscle, Skeletal/innervation
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/physiology
- Necrosis/prevention & control
- Nerve Regeneration/drug effects
- Nerve Regeneration/genetics
- Nerve Regeneration/physiology
- Peptides/administration & dosage
- Peptides/pharmacokinetics
- Peptides/therapeutic use
- Protein Structure, Tertiary/genetics
Collapse
Affiliation(s)
- Daniel P. Reay
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - Michele Yang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - Jon F. Watchko
- Department of Pediatrics, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - Molly Daood
- Department of Pediatrics, Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | | | - Khaleel K. Rehman
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - Denis C. Guttridge
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH, USA, 43210
| | - Paul D. Robbins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| | - Paula R. Clemens
- Neurology Service, Department of Veterans Affairs Medical Center, Pittsburgh, PA, USA, 15240
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA, 15213
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA, 15213
| |
Collapse
|
110
|
Caron AZ, Haroun S, Leblanc E, Trensz F, Guindi C, Amrani A, Grenier G. The proteasome inhibitor MG132 reduces immobilization-induced skeletal muscle atrophy in mice. BMC Musculoskelet Disord 2011; 12:185. [PMID: 21843349 PMCID: PMC3173404 DOI: 10.1186/1471-2474-12-185] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 08/15/2011] [Indexed: 11/10/2022] Open
Abstract
Background Skeletal muscle atrophy is a serious concern for the rehabilitation of patients afflicted by prolonged limb restriction. This debilitating condition is associated with a marked activation of NFκB activity. The ubiquitin-proteasome pathway degrades the NFκB inhibitor IκBα, enabling NFκB to translocate to the nucleus and bind to the target genes that promote muscle atrophy. Although several studies showed that proteasome inhibitors are efficient to reduce atrophy, no studies have demonstrated the ability of these inhibitors to preserve muscle function under catabolic condition. Methods We recently developed a new hindlimb immobilization procedure that induces significant skeletal muscle atrophy and used it to show that an inflammatory process characterized by the up-regulation of TNFα, a known activator of the canonical NFκB pathway, is associated with the atrophy. Here, we used this model to investigate the effect of in vivo proteasome inhibition on the muscle integrity by histological approach. TNFα, IL-1, IL-6, MuRF-1 and Atrogin/MAFbx mRNA level were determined by qPCR. Also, a functional measurement of locomotors activity was performed to determine if the treatment can shorten the rehabilitation period following immobilization. Results In the present study, we showed that the proteasome inhibitor MG132 significantly inhibited IκBα degradation thus preventing NFκB activation in vitro. MG132 preserved muscle and myofiber cross-sectional area by downregulating the muscle-specific ubiquitin ligases atrogin-1/MAFbx and MuRF-1 mRNA in vivo. This effect resulted in a diminished rehabilitation period. Conclusion These finding demonstrate that proteasome inhibitors show potential for the development of pharmacological therapies to prevent muscle atrophy and thus favor muscle rehabilitation.
Collapse
Affiliation(s)
- Annabelle Z Caron
- Centre de Recherche Clinique Étienne-Lebel, 3001-12th Avenue North, Sherbrooke, QC J1H5N4, Canada
| | | | | | | | | | | | | |
Collapse
|
111
|
Radley-Crabb H, Terrill J, Shavlakadze T, Tonkin J, Arthur P, Grounds M. A single 30 min treadmill exercise session is suitable for 'proof-of concept studies' in adult mdx mice: a comparison of the early consequences of two different treadmill protocols. Neuromuscul Disord 2011; 22:170-82. [PMID: 21835619 DOI: 10.1016/j.nmd.2011.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/24/2011] [Accepted: 07/11/2011] [Indexed: 11/15/2022]
Abstract
The extent of muscle pathology in sedentary adult mdx mice is very low and treadmill exercise is often used to increase myofibre necrosis; however, the early events in dystrophic muscle and blood in response to treadmill exercise (leading to myofibre necrosis) are unknown. This study describes in detail two standardised protocols for the treadmill exercise of mdx mice and profiles changes in molecular and cellular events after a single 30 min treadmill session (Protocol A) or after 4 weeks of (twice weekly) treadmill exercise (Protocol B). Both treadmill protocols increased multiple markers of muscle damage. We conclude that a single 30 min treadmill exercise session is a sufficient and conveniently fast screening test and could be used in 'proof-of-concept' studies to evaluate the benefits of pre-clinical drugs in vivo. Myofibre necrosis, blood serum CK and oxidative stress (specifically the ratio of oxidised to reduced protein thiols) are reliable markers of muscle damage after exercise; many parameters demonstrated high biological variation including changes in mRNA levels for key inflammatory cytokines in muscle. The sampling (sacrifice and tissue collection) time after exercise for these parameters is critical. A more precise understanding of the changes in dystrophic muscle after exercise aims to identify biomarkers and new potential therapeutic drug targets for Duchenne Muscular Dystrophy.
Collapse
Affiliation(s)
- Hannah Radley-Crabb
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, Australia.
| | | | | | | | | | | |
Collapse
|
112
|
The soy isoflavone genistein blunts nuclear factor kappa-B, MAPKs and TNF-α activation and ameliorates muscle function and morphology in mdx mice. Neuromuscul Disord 2011; 21:579-89. [DOI: 10.1016/j.nmd.2011.04.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/05/2011] [Accepted: 04/27/2011] [Indexed: 11/18/2022]
|
113
|
Klyen BR, Shavlakadze T, Radley-Crabb HG, Grounds MD, Sampson DD. Identification of muscle necrosis in the mdx mouse model of Duchenne muscular dystrophy using three-dimensional optical coherence tomography. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:076013. [PMID: 21806274 DOI: 10.1117/1.3598842] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Three-dimensional optical coherence tomography (3D-OCT) was used to image the structure and pathology of skeletal muscle tissue from the treadmill-exercised mdx mouse model of human Duchenne muscular dystrophy. Optical coherence tomography (OCT) images of excised muscle samples were compared with co-registered hematoxylin and eosin-stained and Evans blue dye fluorescence histology. We show, for the first time, structural 3D-OCT images of skeletal muscle dystropathology well correlated with co-located histology. OCT could identify morphological features of interest and necrotic lesions within the muscle tissue samples based on intrinsic optical contrast. These findings demonstrate the utility of 3D-OCT for the evaluation of small-animal skeletal muscle morphology and pathology, particularly for studies of mouse models of muscular dystrophy.
Collapse
Affiliation(s)
- Blake R Klyen
- The University of Western Australia, School of Electrical, Electronic and Computer Engineering, Optical+Biomedical Engineering Laboratory, M018, 35 Stirling Highway, Crawley, Western Australia 6009, Australia.
| | | | | | | | | |
Collapse
|
114
|
Cozzoli A, Rolland JF, Capogrosso RF, Sblendorio VT, Longo V, Simonetti S, Nico B, De Luca A. Evaluation of potential synergistic action of a combined treatment with alpha-methyl-prednisolone and taurine on the mdx mouse model of Duchenne muscular dystrophy. Neuropathol Appl Neurobiol 2011; 37:243-56. [PMID: 20618838 DOI: 10.1111/j.1365-2990.2010.01106.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Glucocorticoids are the sole drugs clinically used in Duchenne muscular dystrophy, in spite of the relevant side effects. Combination of glucocorticoids with synergistic drugs may be one strategy to lower doses and control side effects, meanwhile providing wider control of the complex pathology. This study is a preclinical evaluation of the effect of a combined treatment of α-methyl-prednisolone (PDN) with taurine, a safe aminoacid with positive effects on some pathology-related events. METHODS PDN (1 mg/kg/day i.p.) and taurine (1 g/kg/day orally) were administered either alone or in combination, for 4-8 weeks to male dystrophic mdx mice chronically exercised on a treadmill. Effects were assessed in vivo and ex vivo with a variety of methodological approaches. RESULTS In vivo, each treatment significantly increased fore limb strength, a marked synergistic effect being observed with the combination PDN + taurine. Ex vivo, PDN + taurine completely restored the mechanical threshold, an electrophysiological index of calcium homeostasis, of extensor digitorum longus myofibres and the benefit was greater than for PDN alone. In parallel, the overactivity of voltage-independent cation channels in dystrophic myofibres was reduced. No effects were observed on plasma levels of creatine kinase, while lactate dehydrogenase was decreased by taurine and, to a minor extent, by PDN + taurine. A similar histology profile was observed in PDN and PDN + taurine-treated muscles. PDN + taurine significantly increased taurine level in fast-twitch muscle and brain, by high-pressure liquid chromatography analysis. CONCLUSIONS The combination PDN + taurine has additive actions on in vivo and ex vivo functional end points, with less evident advantages on histopathology and biochemical markers of the disease.
Collapse
MESH Headings
- Animals
- Chromatography, High Pressure Liquid
- Creatine Kinase/blood
- Disease Models, Animal
- Drug Synergism
- Drug Therapy, Combination
- Glucocorticoids/administration & dosage
- L-Lactate Dehydrogenase/blood
- Male
- Methylprednisolone/administration & dosage
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle Strength/drug effects
- Muscle, Skeletal/drug effects
- Muscular Dystrophy, Animal/drug therapy
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Patch-Clamp Techniques
- Taurine/administration & dosage
Collapse
Affiliation(s)
- A Cozzoli
- Unit of Pharmacology, Department of Pharmaco-biology, Faculty of Pharmacy Department of Human Anatomy and Histology, Faculty of Medicine, University of Bari Metabolic Disease Unit, Paediatric Hospital Giovanni XXIII, Bari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Potgieter M, Pretorius E, Van der Merwe C, Beukes M, Vieira W, Auer R, Auer M, Meyer S. Histological assessment of SJL/J mice treated with the antioxidants coenzyme Q10 and resveratrol. Micron 2011; 42:275-82. [DOI: 10.1016/j.micron.2010.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Revised: 10/01/2010] [Accepted: 10/03/2010] [Indexed: 01/12/2023]
|
116
|
Gehrig SM, Lynch GS. Emerging drugs for treating skeletal muscle injury and promoting muscle repair. Expert Opin Emerg Drugs 2011; 16:163-82. [DOI: 10.1517/14728214.2010.524743] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
117
|
Nemoto H, Konno S, Sugimoto H, Nakazora H, Nomoto N, Murata M, Kitazono H, Fujioka T. Anti-TNF therapy using etanercept suppresses degenerative and inflammatory changes in skeletal muscle of older SJL/J mice. Exp Mol Pathol 2011; 90:264-70. [PMID: 21324312 DOI: 10.1016/j.yexmp.2011.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Revised: 02/06/2011] [Accepted: 02/07/2011] [Indexed: 12/13/2022]
Abstract
Limb-girdle muscular dystrophy 2B and Miyoshi myopathy are characterized by muscle fiber necrosis caused by a defect in dysferlin and inflammatory changes. SJL/J mice are deficient in dysferlin and display severe inflammatory changes, most notably the presence of cytokines, which may be related to destruction of the sarcolemma. We tested the hypothesis that tumor necrosis factor (TNF) contributes to myofibril necrosis. Administration of etanercept, an agent that blocks TNF, resulted in dose-dependent reductions in inflammatory change, necrosis, and fatty/fibrous change. These findings indicate that TNF does indeed play a role in the damage to muscle in SJL/J mice and that etanercept has the potential to reduce such damage.
Collapse
Affiliation(s)
- Hiroshi Nemoto
- Division of Neurology, Department of Internal Medicine, Toho University Ohashi Medical Center, 2-17-6 Ohashi, Meguro, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
118
|
Dagdeviren S, Kandilci HB, Uysal B, Zeybek ND, Korkusuz P, Gümüsel B, Korkusuz F. Tumor necrosis factor-alpha antagonist administration recovers skeletal muscle dysfunction in ovariectomized rats. J Orthop Res 2011; 29:275-80. [PMID: 20690186 DOI: 10.1002/jor.21226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Skeletal muscles deteriorate after ovariectomy. Molecular pathway of this deterioration has not been defined. Tumor necrosis factor (TNF)-alpha activation is assumed to trigger muscle atrophy and administration of its antagonist is hypothesized to recover this atrophy in rats. Slow-twitch soleus and fast-twitch extensor digitorum longus muscle functions were investigated in intact, ovariectomized (OVX), and OVX plus 10 µg/g/week TNF-alpha antagonist administered female rats. Maximum isometric twitch and tetanic contraction responses were lower in the OVX groups. Maximum isometric twitch amplitudes recovered in the extensor digitorum longus but not in the soleus muscles after TNF-alpha antagonist administration. The decrease in responses to tetanic stimulations recovered in the OVX-TNF group at frequencies higher than 20 Hz in both muscle types. OVX animals body weight was 21% higher than intact animals. Muscle weight to body weight ratios of the OVX groups were higher than the control group which recovered after TNF-alpha antagonist administration. Findings suggest that the functional loss in OVX rat muscles is TNF-alpha pathway dependent. Skeletal muscle atrophy and function after OVX recovered by TNF-alpha antagonist administration.
Collapse
Affiliation(s)
- Sezin Dagdeviren
- Medical Center, Department of Biotechnology, Middle East Technical University, İnönü Bulvarı, Ankara 06531, Turkey
| | | | | | | | | | | | | |
Collapse
|
119
|
Nakagaki WR, Bertran CA, Matsumura CY, Santo-Neto H, Camilli JA. Mechanical, biochemical and morphometric alterations in the femur of mdx mice. Bone 2011; 48:372-9. [PMID: 20850579 DOI: 10.1016/j.bone.2010.09.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 08/20/2010] [Accepted: 09/07/2010] [Indexed: 11/18/2022]
Abstract
The bone tissue abnormalities observed in patients with Duchenne muscular dystrophy are frequently attributed to muscle weakness. In this condition, bones receive fewer mechanical stimuli, compromising the process of bone modeling. In the present study we hypothesize that other factors inherent to the disease might be associated with bone tissue impairment, irrespective of the presence of muscle impairment. Mdx mice lack dystrophin and present cycles of muscle degeneration/regeneration that become more intense in the third week of life. As observed in humans with muscular dystrophy, bone tissue abnormalities were found in mdx mice during more intense muscle degeneration due to age. Under these circumstances, muscle deficit is probably one of the factors promoting these changes. To test our hypothesis, we investigated the changes that occur in the femur of mdx mice at 21 days of age when muscle damage is still not significant. The mechanical (structural and material) and biochemical properties and morphometric characteristics of the femur of mdx and control animals were evaluated. The results demonstrated a lower strength, stiffness and energy absorption capacity in mdx femurs. Higher values for structural (load and stiffness) and material (stress, elastic modulus and toughness) properties were observed in the control group. Mdx femurs were shorter and were characterized by a smaller cortical area and thickness and a smaller area of epiphyseal trabecular bone. The hydroxyproline content was similar in the two groups, but there was a significant difference in the Ca/P ratios. Thermogravimetry showed a higher mineral matrix content in cortical bone of control animals. In conclusion, femurs of mdx mice presented impaired mechanical and biochemical properties as well as changes in collagen organization in the extracellular matrix. Thus, mdx mice developed femoral osteopenia even in the absence of significant muscle fiber degeneration. This weakness of the mdx femur is probably due to genetic factors that are directly or indirectly related to dystrophin deficiency.
Collapse
Affiliation(s)
- Wilson Romero Nakagaki
- Department of Anatomy, Cell Biology and Physiology and Biophysics, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | | | | |
Collapse
|
120
|
Jørgensen LH, Blain A, Greally E, Laval SH, Blamire AM, Davison BJ, Brinkmeier H, MacGowan GA, Schrøder HD, Bushby K, Straub V, Lochmüller H. Long-term blocking of calcium channels in mdx mice results in differential effects on heart and skeletal muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:273-83. [PMID: 21224064 PMCID: PMC3016598 DOI: 10.1016/j.ajpath.2010.11.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 08/19/2010] [Accepted: 09/21/2010] [Indexed: 02/01/2023]
Abstract
The disease mechanisms underlying dystrophin-deficient muscular dystrophy are complex, involving not only muscle membrane fragility, but also dysregulated calcium homeostasis. Specifically, it has been proposed that calcium channels directly initiate a cascade of pathological events by allowing calcium ions to enter the cell. The objective of this study was to investigate the effect of chronically blocking calcium channels with the aminoglycoside antibiotic streptomycin from onset of disease in the mdx mouse model of Duchenne muscular dystrophy (DMD). Treatment in utero onwards delayed onset of dystrophic symptoms in the limb muscle of young mdx mice, but did not prevent degeneration and regeneration events occurring later in the disease course. Long-term treatment had a positive effect on limb muscle pathology, reduced fibrosis, increased sarcolemmal stability, and promoted muscle regeneration in older mice. However, streptomycin treatment did not show positive effects in diaphragm or heart muscle, and heart pathology was worsened. Thus, blocking calcium channels even before disease onset does not prevent dystrophy, making this an unlikely treatment for DMD. These findings highlight the importance of analyzing several time points throughout the life of the treated mice, as well as analyzing many tissues, to get a complete picture of treatment efficacy.
Collapse
Affiliation(s)
- Louise H. Jørgensen
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Alison Blain
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Elizabeth Greally
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Steve H. Laval
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew M. Blamire
- Institute of Cellular Medicine and Newcastle Magnetic Resonance Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Benjamin J. Davison
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Heinrich Brinkmeier
- Institute of Pathophysiology, Ernst Moritz Arndt University of Greifswald, Karlsburg, Germany
| | - Guy A. MacGowan
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Cardiology, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Henrik D. Schrøder
- Department of Clinical Pathology, University of Southern Denmark, Odense C, Denmark
| | - Kate Bushby
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Volker Straub
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Hanns Lochmüller
- Institute of Human Genetics, International Centre of Life, Newcastle University, Newcastle upon Tyne, United Kingdom
- Address reprint requests to Professor Hanns Lochmüller, MD, Institute of Human Genetics, International Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| |
Collapse
|
121
|
Piers AT, Lavin T, Radley-Crabb HG, Bakker AJ, Grounds MD, Pinniger GJ. Blockade of TNF in vivo using cV1q antibody reduces contractile dysfunction of skeletal muscle in response to eccentric exercise in dystrophic mdx and normal mice. Neuromuscul Disord 2010; 21:132-41. [PMID: 21055937 DOI: 10.1016/j.nmd.2010.09.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 09/22/2010] [Accepted: 09/23/2010] [Indexed: 01/25/2023]
Abstract
This study evaluated the contribution of the pro-inflammatory cytokine, tumour necrosis factor (TNF) to the severity of exercise-induced muscle damage and subsequent myofibre necrosis in mdx mice. Adult mdx and non-dystrophic C57 mice were treated with the mouse-specific TNF antibody cV1q before undergoing a damaging eccentric contraction protocol performed in vivo on a custom built mouse dynamometer. Muscle damage was quantified by (i) contractile dysfunction (initial torque deficit) immediately after the protocol, (ii) subsequent myofibre necrosis 48 h later. Blockade of TNF using cV1q significantly reduced contractile dysfunction in mdx and C57 mice compared with mice injected with the negative control antibody (cVaM) and un-treated mice. Furthermore, cV1q treatment significantly reduced myofibre necrosis in mdx mice. This in vivo evidence that cV1q reduces the TNF-mediated adverse response to exercise-induced muscle damage supports the use of targeted anti-TNF treatments to reduce the severity of the functional deficit and dystropathology in DMD.
Collapse
Affiliation(s)
- A T Piers
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | | | | | | | | | | |
Collapse
|
122
|
Van Erp C, Loch D, Laws N, Trebbin A, Hoey AJ. Timeline of cardiac dystrophy in 3-18-month-old MDX mice. Muscle Nerve 2010; 42:504-13. [PMID: 20589894 DOI: 10.1002/mus.21716] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The dystrophin-deficient (mdx) mouse remains the most commonly used model for Duchenne muscular dystrophy (DMD). Mdx mice show a predominantly covert cardiomyopathy, the hallmark of which is fibrosis. We compared mdx and normal mice at six ages (3, 6, 9, 12, 15, and 18 months) using in vivo assessment of cardiac function, selective collagen staining, and measures of TGF-β mRNA, Evans blue dye infiltration, macrophage infiltration, and aortic wall thickness. Clear temporal progression was demonstrated, including early fragility of cardiomyocyte membranes, which has an unrelated impact on cardiac function but is associated with macrophage infiltration and fibrosis. Aortic wall thickness is less in older mdx mice. Mdx mice display impaired responses to inotropic challenge from a young age; this is indicative of altered adrenoreceptor function. We draw attention to the paradox of ongoing fibrosis in mdx hearts without a strong molecular signature (in the form of TGF-β mRNA expression).
Collapse
Affiliation(s)
- Christel Van Erp
- Centre for Systems Biology, Faculty of Sciences, University of Southern Queensland, Toowoomba, Queensland, Australia
| | | | | | | | | |
Collapse
|
123
|
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
|
124
|
Iannitti T, Capone S, Feder D, Palmieri B. Clinical use of immunosuppressants in Duchenne muscular dystrophy. J Clin Neuromuscul Dis 2010; 12:1-21. [PMID: 20808159 DOI: 10.1097/cnd.0b013e3181d4a4f9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a degenerative disease primarily affecting voluntary muscles with secondary consequences on heart and breathing muscles. DMD is an X-linked recessive disease that results in the loss of dystrophin, a key muscle protein. Inflammation can play different roles in DMD; it can be a secondary response to muscle degeneration, a primary cause of degeneration, or can contribute to the disease progression. Several immunosuppressants have been used with the aim to reduce the inflammation associated with DMD. Most recently, myoblast transplantation has shown the possibility to restore the dystrophin lack in the DMD patient's muscle fibers and this evidence has emphasized the importance of the use of immunosuppressants and the necessity of studying them and their secondary effects. The aim of this review is to analyze the main immunosuppressants drugs starting from the mdx mice experiments and concluding with the most recent human clinical studies.
Collapse
Affiliation(s)
- Tommaso Iannitti
- Department of Biological and Biomedical Sciences, School of Life Sciences, Glasgow Caledonian University, Glasgow, UK.
| | | | | | | |
Collapse
|
125
|
TNF inhibits Notch-1 in skeletal muscle cells by Ezh2 and DNA methylation mediated repression: implications in duchenne muscular dystrophy. PLoS One 2010; 5:e12479. [PMID: 20814569 PMCID: PMC2930001 DOI: 10.1371/journal.pone.0012479] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 08/01/2010] [Indexed: 12/21/2022] Open
Abstract
Background Classical NF-κB signaling functions as a negative regulator of skeletal myogenesis through potentially multiple mechanisms. The inhibitory actions of TNFα on skeletal muscle differentiation are mediated in part through sustained NF-κB activity. In dystrophic muscles, NF-κB activity is compartmentalized to myofibers to inhibit regeneration by limiting the number of myogenic progenitor cells. This regulation coincides with elevated levels of muscle derived TNFα that is also under IKKβ and NF-κB control. Methodology/Principal Findings Based on these findings we speculated that in DMD, TNFα secreted from myotubes inhibits regeneration by directly acting on satellite cells. Analysis of several satellite cell regulators revealed that TNFα is capable of inhibiting Notch-1 in satellite cells and C2C12 myoblasts, which was also found to be dependent on NF-κB. Notch-1 inhibition occurred at the mRNA level suggesting a transcriptional repression mechanism. Unlike its classical mode of action, TNFα stimulated the recruitment of Ezh2 and Dnmt-3b to coordinate histone and DNA methylation, respectively. Dnmt-3b recruitment was dependent on Ezh2. Conclusions/Significance We propose that in dystrophic muscles, elevated levels of TNFα and NF-κB inhibit the regenerative potential of satellite cells via epigenetic silencing of the Notch-1 gene.
Collapse
|
126
|
Leite PEC, Lagrota-Candido J, Moraes L, D'Elia L, Pinheiro DF, da Silva RF, Yamasaki EN, Quirico-Santos T. Nicotinic acetylcholine receptor activation reduces skeletal muscle inflammation of mdx mice. J Neuroimmunol 2010; 227:44-51. [PMID: 20615555 DOI: 10.1016/j.jneuroim.2010.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 06/02/2010] [Accepted: 06/03/2010] [Indexed: 11/29/2022]
Abstract
Mdx mice develop an inflammatory myopathy characterized at different ages by myonecrosis with scattered inflammatory infiltrates followed by muscular regeneration and later persistent fibrosis. This work aimed to verify the putative anti-inflammatory role of nicotinic acetylcholine receptor (nAChR) in the mdx muscular lesion. Mitigation of myonecrosis and decreased TNFα production were accompanied by increased numbers of F4/80 macrophages expressing nAChRα7. In vivo treatment with nicotine attenuated muscular inflammation characterized by reduced metalloprotease MMP-9 activity, TNFα and NFkB content and increased muscular regeneration. Our data indicate that nAChR activation influences local inflammatory responses in the muscular lesion of mdx mice.
Collapse
Affiliation(s)
- Paulo Emílio Corrêa Leite
- Laboratório de Patologia Celular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Soluble TNF-α receptor secretion from healthy or dystrophic mice after AAV6-mediated muscle gene transfer. Gene Ther 2010; 17:1400-10. [PMID: 20596058 DOI: 10.1038/gt.2010.94] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Muscle is an attractive target because it is easily accessible; it also offers a permissive environment for adeno-associated virus (AAV)-mediated gene transfer and has an abundant blood vascular supply providing an efficient transport system for the secretion of proteins. However, gene therapy of dystrophic muscle may be more difficult than that of healthy tissue because of degenerative-regenerative processes, and also because of the inflammatory context. In this study we followed the expression levels of secreted inhibitors of the proinflammatory tumor necrosis factor (TNF) cytokine after intramuscular (i.m.) injection of AAV6 into dystrophic mdx and healthy C57BL/10 mice. We used two chimeric proteins, namely, the human or murine TNF-soluble receptor I fused with the murine heavy immunoglobulin chain. We conducted an AAV6 dose-response study and determined the kinetics of transgene expression. In addition, we followed the antibody response against the transgenes and studied their expression pattern in the muscle. Our results show that transduction efficiency is reduced in dystrophic muscles as compared with healthy ones. Furthermore, we found that the immune response against the secreted protein is stronger in mdx mice. Together, our results underscore that the pathological state of the muscle has to be taken into consideration when designing gene therapy approaches.
Collapse
|
128
|
Sciorati C, Buono R, Azzoni E, Casati S, Ciuffreda P, D'Angelo G, Cattaneo D, Brunelli S, Clementi E. Co-administration of ibuprofen and nitric oxide is an effective experimental therapy for muscular dystrophy, with immediate applicability to humans. Br J Pharmacol 2010; 160:1550-60. [PMID: 20590643 PMCID: PMC2938824 DOI: 10.1111/j.1476-5381.2010.00809.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/08/2010] [Accepted: 03/11/2010] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND AND PURPOSE Current therapies for muscular dystrophy are based on corticosteroids. Significant side effects associated with these therapies have prompted several studies aimed at identifying possible alternative strategies. As inflammation and defects of nitric oxide (NO) generation are key pathogenic events in muscular dystrophies, we have studied the effects of combining the NO donor isosorbide dinitrate (ISDN) and the non-steroidal anti-inflammatory drug ibuprofen. EXPERIMENTAL APPROACH alpha-Sarcoglycan-null mice were treated for up to 8 months with ISDN (30 mg.kg(-1)) plus ibuprofen (50 mg.kg(-1)) administered daily in the diet. Effects of ISDN and ibuprofen alone were assessed in parallel. Drug effects on animal motility and muscle function, muscle damage, inflammatory infiltrates and cytokine levels, as well as muscle regeneration including assessment of endogenous stem cell pool, were measured at selected time points. KEY RESULTS Combination of ibuprofen and ISDN stimulated regeneration capacity, of myogenic precursor cells, reduced muscle necrotic damage and inflammation. Muscle function in terms of free voluntary movement and resistance to exercise was maintained throughout the time window analysed. The effects of ISDN and ibuprofen administered separately were transient and significantly lower than those induced by their combination. CONCLUSIONS AND IMPLICATIONS Co-administration of NO and ibuprofen provided synergistic beneficial effects in a mouse model of muscular dystrophy, leading to an effective therapy. Our results open the possibility of immediate clinical testing of a combination of ISDN and ibuprofen in dystrophic patients, as both components are approved for use in humans, with a good safety profile.
Collapse
Affiliation(s)
- Clara Sciorati
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
| | - Roberta Buono
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Emanuele Azzoni
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
- Department of Experimental Medicine, University of Milano-BicoccaMonza, Italy
| | - Silvana Casati
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Pierangela Ciuffreda
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | | | - Dario Cattaneo
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
| | - Silvia Brunelli
- San Raffaele Scientific Institute, Stem Cell Research InstituteMilan, Italy
- Department of Experimental Medicine, University of Milano-BicoccaMonza, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Preclinical Sciences, University Hospital ‘Luigi Sacco’, Università di MilanoMilan, Italy,
- E. Medea Scientific InstituteBosisio Parini, Italy
| |
Collapse
|
129
|
Chahbouni M, Escames G, Venegas C, Sevilla B, García JA, López LC, Muñoz-Hoyos A, Molina-Carballo A, Acuña-Castroviejo D. Melatonin treatment normalizes plasma pro-inflammatory cytokines and nitrosative/oxidative stress in patients suffering from Duchenne muscular dystrophy. J Pineal Res 2010; 48:282-289. [PMID: 20210854 DOI: 10.1111/j.1600-079x.2010.00752.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD), a lethal disorder characterized by dystrophin absence, courses with chronic inflammation, sarcolemmal damage, and skeletal muscle degeneration. Among the multiple pathogenic mechanisms proposed for DMD, oxidative stress and inflammation are directly involved in the dystrophic process. Unfortunately, there is no current treatment for DMD, and the inflammatory process is an important target for therapies. Based on the antioxidant and anti-inflammatory properties of melatonin, we investigated whether melatonin treatment may reduce the dystrophic process. Ten DMD patients aged 12.8 +/- 0.98 yr, were treated with melatonin (60 mg at 21:00 hr plus 10 mg at 09:00 hr), and plasma levels of lipid peroxidation (LPO), nitrites (NO(x)), interleukin (IL)-1beta, IL-2, IL-6, tumor necrosis factor-alpha, interferon-gamma, and plasma markers of muscle injury, were determined at 3, 6 and 9 months of treatment. Healthy age- and sex-matched subjects were used as controls. The results show a significant increase in LPO, NO(x), and cytokine levels in plasma of DMD patients compared with controls. Melatonin administration reduced these values to control levels at 3 months of treatment, decreasing further 9 months later. In parallel, melatonin also reduced plasma levels of creatine kinase (CK; 50%), lactate dehydrogenase (28%), aspartate aminotransferase (28%), alanine aminotransferase (20%), and myoglobin (13%). These findings strongly support the conclusion that melatonin administration significantly reduced the hyperoxidative and inflammatory process in DMD patients, reducing the muscle degenerative process.
Collapse
Affiliation(s)
- Mariam Chahbouni
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Carmen Venegas
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Belén Sevilla
- Unidad de Gestión Clínica de Pediatría, Hospital Universitario San Cecilio, Granada, Spain
| | - José Antonio García
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Luis C López
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Antonio Muñoz-Hoyos
- Unidad de Gestión Clínica de Pediatría, Hospital Universitario San Cecilio, Granada, Spain
| | | | - Darío Acuña-Castroviejo
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Servicio de Análisis Clínicos, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
130
|
Ichim TE, Alexandrescu DT, Solano F, Lara F, Campion RDN, Paris E, Woods EJ, Murphy MP, Dasanu CA, Patel AN, Marleau AM, Leal A, Riordan NH. Mesenchymal stem cells as anti-inflammatories: implications for treatment of Duchenne muscular dystrophy. Cell Immunol 2010; 260:75-82. [PMID: 19917503 DOI: 10.1016/j.cellimm.2009.10.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 10/13/2009] [Indexed: 01/01/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked musculodegenerative condition consisting of an underlying genetic defect whose manifestation is augmented by inflammatory mechanisms. Previous treatment approaches using gene replacement, exon-skipping or allogeneic cell therapy have been relatively unsuccessful. The only intervention to mediate improvement in survival, albeit minor, is glucocorticoid treatment. Given this modality appears to function via suppression of underlying inflammation; we focus this review on the inflammatory response as a target for mesenchymal stem cell (MSC) therapy. In contrast to other cell based therapies attempted in DMD, MSC have the advantages of (a) ability to fuse with and genetically complement dystrophic muscle; (b) possess anti-inflammatory activities; and (c) produce trophic factors that may augment activity of endogenous repair cells. We conclude by describing one practical scenario of stem cell therapy for DMD.
Collapse
|
131
|
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
|
132
|
Evans NP, Call JA, Bassaganya-Riera J, Robertson JL, Grange RW. Green tea extract decreases muscle pathology and NF-kappaB immunostaining in regenerating muscle fibers of mdx mice. Clin Nutr 2009; 29:391-8. [PMID: 19897286 DOI: 10.1016/j.clnu.2009.10.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 08/09/2009] [Accepted: 10/08/2009] [Indexed: 11/17/2022]
Abstract
BACKGROUND & AIMS Duchenne muscular dystrophy is a debilitating genetic disorder characterized by severe muscle wasting and early death in afflicted boys. The primary cause of this disease is mutations in the dystrophin gene resulting in massive muscle degeneration and inflammation. The purpose of this study was to determine if dystrophic muscle pathology and inflammation were decreased by pre-natal and early dietary intervention with green tea extract. METHODS Mdx breeder mice and pups were fed diets containing 0.25% or 0.5% green tea extract and compared to untreated mdx and C57BL/6J mice. Serum creatine kinase was assessed as a systemic indicator of muscle damage. Quantitative histopathological and immunohistochemical techniques were used to determine muscle pathology, macrophage infiltration, and NF-kappaB localization. RESULTS Early treatment of mdx mice with green tea extract significantly decreased serum creatine kinase by approximately 85% at age 42 days (P< or =0.05). In these mice, the area of normal fiber morphology was increased by as much as approximately 32% (P< or =0.05). The primary histopathological change was a approximately 21% decrease in the area of regenerating fibers (P< or =0.05). NF-kappaB staining in regenerating muscle fibers was also significantly decreased in green tea extract-treated mdx mice when compared to untreated mdx mice (P< or =0.05). CONCLUSION Early treatment with green tea extract decreases dystrophic muscle pathology potentially by regulating NF-kappaB activity in regenerating muscle fibers.
Collapse
MESH Headings
- Aging
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Antioxidants/pharmacology
- Antioxidants/therapeutic use
- Biomarkers/blood
- Dose-Response Relationship, Drug
- Female
- Macrophages/drug effects
- Male
- Mice
- Mice, Inbred mdx
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/blood
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/prevention & control
- NF-kappa B/metabolism
- Necrosis/pathology
- Necrosis/prevention & control
- Phytotherapy
- Plant Extracts/pharmacology
- Plant Extracts/therapeutic use
- Pregnancy
- Prenatal Exposure Delayed Effects
- Random Allocation
- Regeneration/drug effects
- Tea/chemistry
Collapse
Affiliation(s)
- Nicholas P Evans
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, 338 Wallace Hall, Blacksburg, VA 24061, USA.
| | | | | | | | | |
Collapse
|
133
|
Use of pifithrin to inhibit p53-mediated signalling of TNF in dystrophic muscles of mdx mice. Mol Cell Biochem 2009; 337:119-31. [PMID: 19859789 DOI: 10.1007/s11010-009-0291-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 10/08/2009] [Indexed: 10/20/2022]
Abstract
Tumour Necrosis Factor (TNF) plays a major role in exacerbating necrosis of dystrophic muscle; however, the precise molecular mechanism underlying this effect of TNF is unknown. This study investigates the role that p53 plays in TNF-mediated necrosis of dystrophic myofibres by inhibiting p53 using pifithrin-alpha and three pifithrin-beta analogues. Tissue culture studies using C2C12 myoblasts established that pifithrin-alpha was toxic to differentiating myoblasts at concentrations greater than 10 muM. While non-toxic concentrations of pifithrin-alpha did not prevent the TNF-mediated inhibition of myoblast differentiation, Western blots indicated that nuclear levels of p53 were higher in TNF-treated myoblasts indicating that TNF does elevate p53. In contrast, in vivo studies in adult mdx mice showed that pifithrin-alpha significantly reduced myofibre necrosis that resulted from voluntary wheel running over 48 h. These results support the hypothesis that p53 plays some role in TNF-mediated necrosis of dystrophic muscle and present a potential new target for therapeutic interventions.
Collapse
|
134
|
Messina S, Bitto A, Aguennouz M, Mazzeo A, Migliorato A, Polito F, Irrera N, Altavilla D, Vita GL, Russo M, Naro A, De Pasquale MG, Rizzuto E, Musarò A, Squadrito F, Vita G. Flavocoxid counteracts muscle necrosis and improves functional properties in mdx mice: a comparison study with methylprednisolone. Exp Neurol 2009; 220:349-58. [PMID: 19786019 DOI: 10.1016/j.expneurol.2009.09.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/18/2009] [Accepted: 09/19/2009] [Indexed: 10/20/2022]
Abstract
Muscle degeneration in dystrophic muscle is exacerbated by the endogenous inflammatory response and increased oxidative stress. A key role is played by nuclear factor(NF)-kappaB. We showed that NF-kappaB inhibition through compounds with also antioxidant properties has beneficial effects in mdx mice, the murine model of Duchenne muscular dystrophy (DMD), but these drugs are not available for clinical studies. We evaluated whether flavocoxid, a mixed flavonoid extract with anti-inflammatory, antioxidant and NF-kappaB inhibiting properties, has beneficial effects in mdx mice in comparison with methylprednisolone, the gold standard treatment for DMD patients. Five-week-old mdx mice were treated for 5 weeks with flavocoxid, methylprednisolone or vehicle. The evaluation of in vivo and ex vivo functional properties and morphological parameters was performed. Serum samples were assayed for oxidative stress markers, creatine-kinase (CK) and leukotriene B-4. Cyclooxygenase-2 (COX-2), 5-lipoxygenase (5-LOX), tumor necrosis factor-alpha, p-38, JNK1 expression was evaluated in muscle by western blot analysis. NF-kappaB binding activity was investigated by electrophoresis mobility shift assay. The administration of flavocoxid: (1) ameliorated functional properties in vivo and ex vivo; (2) reduced CK; (3) reduced the expression of oxidative stress markers and of inflammatory mediators; (4) inhibited NF-kappaB and mitogen-activated protein kinases (MAPKs) signal pathways; (5) reduced muscle necrosis and enhanced regeneration. Our results highlight the detrimental effects of oxidative stress and NF-kappaB, MAPKs and COX/5-LOX pathways in the dystrophic process and show that flavocoxid is more effective in mdx mice than methylprednisolone.
Collapse
Affiliation(s)
- Sonia Messina
- Department of Neurosciences, Psychiatry and Anaesthesiology, Unit of Neurology and Neuromuscular Diseases, AOU Policlinico G. Martino, 98125 Messina, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Wallace GQ, McNally EM. Mechanisms of muscle degeneration, regeneration, and repair in the muscular dystrophies. Annu Rev Physiol 2009; 71:37-57. [PMID: 18808326 DOI: 10.1146/annurev.physiol.010908.163216] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To withstand the rigors of contraction, muscle fibers have specialized protein complexes that buffer against mechanical stress and a multifaceted repair system that is rapidly activated after injury. Genetic studies first identified the mechanosensory signaling network that connects the structural elements of muscle and, more recently, have identified repair elements of muscle. Defects in the genes encoding the components of these systems lead to muscular dystrophy, a family of genetic disorders characterized by progressive muscle wasting. Although the age of onset, affected muscles, and severity vary considerably, all muscular dystrophies are characterized by muscle necrosis that overtakes the regenerative capacity of muscle. The resulting replacement of muscle by fatty and fibrous tissue leaves muscle increasingly weak and nonfunctional. This review discusses the cellular mechanisms that are primarily and secondarily disrupted in muscular dystrophy, focusing on membrane degeneration, muscle regeneration, and the repair of muscle.
Collapse
Affiliation(s)
- Gregory Q Wallace
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
136
|
Vercherat C, Chung TK, Yalcin S, Gulbagci N, Gopinadhan S, Ghaffari S, Taneja R. Stra13 regulates oxidative stress mediated skeletal muscle degeneration. Hum Mol Genet 2009; 18:4304-16. [DOI: 10.1093/hmg/ddp383] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
137
|
Huang P, Zhao XS, Fields M, Ransohoff RM, Zhou L. Imatinib attenuates skeletal muscle dystrophy in mdx mice. FASEB J 2009; 23:2539-48. [PMID: 19289603 PMCID: PMC2717779 DOI: 10.1096/fj.09-129833] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 02/19/2009] [Indexed: 01/19/2023]
Abstract
Duchenne-Meryon muscular dystrophy (DMD) is the most common and lethal genetic muscle disease. Ameliorating muscle necrosis, inflammation, and fibrosis represents an important therapeutic approach for DMD. Imatinib, an antineoplastic agent, demonstrated antiinflammatory and antifibrotic effects in liver, kidney, lung, and skin of various animal models. This study tested antiinflammatory and antifibrotic effects of imatinib in mdx mice, a DMD mouse model. We treated mdx mice with intraperitoneal injections of imatinib at the peak of limb muscle inflammation and the onset of diaphragm fibrosis. Controls received PBS vehicle or were left untreated. Muscle necrosis, inflammation, fibrosis, and function were evaluated by measuring serum CK activity, endomysial CD45 immunoreactive inflammation area, endomysial collagen III deposition, and hind limb grip strength. Phosphorylation of the tyrosine kinase targets of imatinib was assessed by Western blot in diaphragm tissue and in primary cultures of peritoneal macrophages and skeletal muscle fibroblasts. Imatinib markedly reduced muscle necrosis, inflammation, and fibrosis, and significantly improved hind limb grip strength in mdx mice. Reduced clinical disease was accompanied by inhibition of c-abl and PDGFR phosphorylation and suppression of TNF-alpha and IL-1beta expression. Imatinib therapy for DMD may hold promise for ameliorating muscle necrosis, inflammation, and fibrosis by inhibiting c-abl and PDGFR signaling pathways and downstream inflammatory cytokine and fibrotic gene expression.
Collapse
MESH Headings
- Animals
- Base Sequence
- Benzamides
- Creatine Kinase/blood
- Diaphragm/drug effects
- Diaphragm/pathology
- Gene Expression/drug effects
- Imatinib Mesylate
- In Vitro Techniques
- Inflammation/drug therapy
- Inflammation/pathology
- Interleukin-1beta/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle Strength/drug effects
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Animal/drug therapy
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/physiopathology
- Necrosis
- Phosphorylation
- Piperazines/pharmacology
- Proto-Oncogene Proteins c-abl/metabolism
- Pyrimidines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Smad2 Protein/metabolism
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Ping Huang
- Department of Neurology/Neurological Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195 USA
| | | | | | | | | |
Collapse
|
138
|
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
|
139
|
Modulation of caspase activity regulates skeletal muscle regeneration and function in response to vasopressin and tumor necrosis factor. PLoS One 2009; 4:e5570. [PMID: 19440308 PMCID: PMC2680623 DOI: 10.1371/journal.pone.0005570] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Accepted: 04/20/2009] [Indexed: 11/19/2022] Open
Abstract
Muscle homeostasis involves de novo myogenesis, as observed in conditions of acute or chronic muscle damage. Tumor Necrosis Factor (TNF) triggers skeletal muscle wasting in several pathological conditions and inhibits muscle regeneration. We show that intramuscular treatment with the myogenic factor Arg8-vasopressin (AVP) enhanced skeletal muscle regeneration and rescued the inhibitory effects of TNF on muscle regeneration. The functional analysis of regenerating muscle performance following TNF or AVP treatments revealed that these factors exerted opposite effects on muscle function. Principal component analysis showed that TNF and AVP mainly affect muscle tetanic force and fatigue. Importantly, AVP counteracted the effects of TNF on muscle function when delivered in combination with the latter. Muscle regeneration is, at least in part, regulated by caspase activation, and AVP abrogated TNF-dependent caspase activation. The contrasting effects of AVP and TNF in vivo are recapitulated in myogenic cell cultures, which express both PW1, a caspase activator, and Hsp70, a caspase inhibitor. We identified PW1 as a potential Hsp70 partner by screening for proteins interacting with PW1. Hsp70 and PW1 co-immunoprecipitated and co-localized in muscle cells. In vivo Hsp70 protein level was upregulated by AVP, and Hsp70 overexpression counteracted the TNF block of muscle regeneration. Our results show that AVP counteracts the effects of TNF through cross-talk at the Hsp70 level. Therefore, muscle regeneration, both in the absence and in the presence of cytokines may be enhanced by increasing Hsp70 expression.
Collapse
|
140
|
Dorchies OM, Wagner S, Buetler TM, Ruegg UT. Protection of dystrophic muscle cells with polyphenols from green tea correlates with improved glutathione balance and increased expression of 67LR, a receptor for (-)-epigallocatechin gallate. Biofactors 2009; 35:279-94. [PMID: 19322813 DOI: 10.1002/biof.34] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by the absence of the protein dystrophin. Because oxidative stress contributes to the pathogenesis of DMD, we investigated if a green tea polyphenol blend (GTP) and its major polyphenol (-)-epigallocatechin gallate (EGCg), could protect muscle cell primary cultures from oxidative damage induced by hydrogen peroxide (H(2)O(2)) in the widely used mdx mouse model. On-line fluorimetric measurements using an H(2)O(2)-sensitive probe indicated that GTP and EGCg scavenged peroxide in a concentration-dependent manner. A 48 h exposure to EGCg increased glutathione content but did not alter the expression of proteins involved in membrane stabilization and repair. Pretreatment of dystrophic cultures with GTP or EGCg 48 h before exposure to H(2)O(2) improved cell survival. Normal cultures were protected by GTP but not by EGCg. 67LR, a receptor for EGCg, was seven times more abundant in dystrophic compared with normal cultures. Altogether our results demonstrate that GTP and EGCg protect muscle cells by scavenging H(2)O(2) and by improving the glutathione balance. In addition, the higher levels of 67LR in dystrophic muscle cells compared with normal ones likely contribute to EGCg-mediated survival.
Collapse
Affiliation(s)
- Olivier M Dorchies
- Laboratory of Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | | | | | | |
Collapse
|
141
|
Burdi R, Rolland JF, Fraysse B, Litvinova K, Cozzoli A, Giannuzzi V, Liantonio A, Camerino GM, Sblendorio V, Capogrosso RF, Palmieri B, Andreetta F, Confalonieri P, De Benedictis L, Montagnani M, De Luca A. Multiple pathological events in exercised dystrophic mdx mice are targeted by pentoxifylline: outcome of a large array of in vivo and ex vivo tests. J Appl Physiol (1985) 2009; 106:1311-24. [PMID: 19131478 DOI: 10.1152/japplphysiol.90985.2008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The phosphodiesterases inhibitor pentoxifylline gained attention for Duchenne muscular dystrophy therapy for its claimed anti-inflammatory, antioxidant, and antifibrotic action. A recent finding also showed that pentoxifylline counteracts the abnormal overactivity of a voltage-independent calcium channel in myofibers of dystrophic mdx mice. The possible link between workload, altered calcium homeostasis, and oxidative stress pushed toward a more detailed investigation. Thus a 4- to 8-wk treatment with pentoxifylline (50 mg x kg(-1) x day(-1) ip) was performed in mdx mice, undergoing or not a chronic exercise on treadmill. In vivo, the treatment partially increased forelimb strength and enhanced resistance to treadmill running in exercised animals. Ex vivo, pentoxifylline restored the mechanical threshold, an electrophysiological index of calcium homeostasis, and reduced resting cytosolic calcium in extensor digitorum longus muscle fibers. Mn quenching and patch-clamp technique confirmed that this effect was paralleled by a drug-induced reduction of membrane permeability to calcium. The treatment also significantly enhanced isometric tetanic tension in mdx diaphragm. The plasma levels of creatine kinase and reactive oxygen species were both significantly reduced in treated-exercised animals. Dihydroethidium staining, used as an indicator of reactive oxygen species production, showed that pentoxifylline significantly reduced the exercise-induced increase in fluorescence in the mdx tibialis anterior muscle. A significant decrease in connective tissue area and profibrotic cytokine transforming growth factor-beta(1) was solely found in tibialis anterior muscle. In both diaphragm and gastrocnemius muscle, a significant increase in neural cell adhesion molecule-positive area was instead observed. This data supports the interest toward pentoxifylline and allows insight in the level of cross talk between pathogenetic events in workloaded dystrophic muscle.
Collapse
Affiliation(s)
- Rosa Burdi
- Unit of Pharmacology, Department of Pharmaco-biology, Faculty of Pharmacy, University of Bari, Bari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Willmann R, Possekel S, Dubach-Powell J, Meier T, Ruegg MA. Mammalian animal models for Duchenne muscular dystrophy. Neuromuscul Disord 2009; 19:241-9. [PMID: 19217290 DOI: 10.1016/j.nmd.2008.11.015] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 11/24/2008] [Accepted: 11/27/2008] [Indexed: 11/27/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disease that affects boys and leads to early death. In the quest for new treatments that improve the quality of life and in the search for a possible definitive cure, the use of animal models plays undoubtedly an important role. Therefore, a number of different mammalian models for DMD have been described. Much knowledge on the molecular mechanisms underlying the disease has arisen from studies in these animals. However, the use of different models does not often allow a direct comparison of results obtained in preclinical trials and therefore hinders a straightforward translational research. In the frame of "TREAT-NMD", a European Network of Excellence addressing the fragmentation in the assessment and treatment of neuromuscular diseases, we compare here the currently used mammalian animal models for DMD with the aim of selecting and recommending the most appropriate ones for preclinical efficacy testing of new therapeutic strategies.
Collapse
Affiliation(s)
- Raffaella Willmann
- Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland
| | | | | | | | | |
Collapse
|
143
|
Laure L, Suel L, Roudaut C, Bourg N, Ouali A, Bartoli M, Richard I, Danièle N. Cardiac ankyrin repeat protein is a marker of skeletal muscle pathological remodelling. FEBS J 2009; 276:669-84. [PMID: 19143834 DOI: 10.1111/j.1742-4658.2008.06814.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In an attempt to identify potential therapeutic targets for the correction of muscle wasting, the gene expression of several pivotal proteins involved in protein metabolism was investigated in experimental atrophy induced by transient or definitive denervation, as well as in four animal models of muscular dystrophies (deficient for calpain 3, dysferlin, alpha-sarcoglycan and dystrophin, respectively). The results showed that: (a) the components of the ubiquitin-proteasome pathway are upregulated during the very early phases of atrophy but do not greatly increase in the muscular dystrophy models; (b) forkhead box protein O1 mRNA expression is augmented in the muscles of a limb girdle muscular dystrophy 2A murine model; and (c) the expression of cardiac ankyrin repeat protein (CARP), a regulator of transcription factors, appears to be persistently upregulated in every condition, suggesting that CARP could be a hub protein participating in common pathological molecular pathway(s). Interestingly, the mRNA level of a cell cycle inhibitor known to be upregulated by CARP in other tissues, p21(WAF1/CIP1), is consistently increased whenever CARP is upregulated. CARP overexpression in muscle fibres fails to affect their calibre, indicating that CARP per se cannot initiate atrophy. However, a switch towards fast-twitch fibres is observed, suggesting that CARP plays a role in skeletal muscle plasticity. The observation that p21(WAF1/CIP1) is upregulated, put in perspective with the effects of CARP on the fibre type, fits well with the idea that the mechanisms at stake might be required to oppose muscle remodelling in skeletal muscle.
Collapse
|
144
|
Matsumura CY, Pertille A, Albuquerque TC, Santo Neto H, Marques MJ. Diltiazem and verapamil protect dystrophin-deficient muscle fibers ofMDXmice from degeneration: A potential role in calcium buffering and sarcolemmal stability. Muscle Nerve 2009; 39:167-76. [DOI: 10.1002/mus.21188] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
145
|
Abstract
The muscular dystrophies are a group of genetically and phenotypically heterogeneously inherited diseases characterized by progressive muscle wasting, which can lead to premature death in severe forms such as Duchenne muscular dystrophy (DMD). In many cases they are caused by the absence of proteins that are critical components of the dystrophin-glycoprotein complex, which links the cytoskeleton and the basal lamina. There is no effective treatment for these disorders at present, but several novel strategies for replacing or repairing the defective gene are in development, with early encouraging results from animal models. We review these strategies, which include the use of stem cells of different tissue origins, gene replacement therapies mediated by various viral vectors, and transcript repair treatments using exon skipping strategies. We comment on their advantages and on limitations that must be overcome before successful application to human patients. Our focus is on studies in a clinically relevant large canine model of DMD. Recent advances in the field suggest that effective therapies for muscular dystrophies are on the horizon. Because of the complex nature of these diseases, it may be necessary to combine multiple approaches to achieve a successful treatment.
Collapse
Affiliation(s)
- Zejing Wang
- Division of Clinical Research, Fred Hutchinson Cancer Research Center in Seattle, Washington 98109, USA
| | | | | | | |
Collapse
|
146
|
Walters KB, Dodd ME, Mathias JR, Gallagher AJ, Bennin DA, Rhodes J, Kanki JP, Look AT, Grinblat Y, Huttenlocher A. Muscle degeneration and leukocyte infiltration caused by mutation of zebrafish Fad24. Dev Dyn 2009; 238:86-99. [PMID: 19097072 PMCID: PMC2843540 DOI: 10.1002/dvdy.21821] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Factor for adipocyte differentiation 24 (fad24) is a novel gene that has been implicated in adipocyte differentiation and DNA replication. In a screen for zebrafish mutants that have an abnormal tissue distribution of neutrophils, we identified an insertional allele of fad24, fad24hi1019. Homozygous fad24hi1019 larvae exhibit muscle degeneration accompanied by leukocyte infiltration. Muscle degeneration was extensive and included tissue apoptosis and disorganized, poorly striated muscle fibers. Blocking apoptosis using pan-caspase inhibitors resulted in decreased neutrophil recruitment into the body of the larva, suggesting a causative link between apoptosis and leukocyte infiltration. These findings suggest that zebrafish is a powerful genetic model system to address the interplay between muscle degeneration and leukocyte infiltration, and indicate that tissue apoptosis may contribute to neutrophil recruitment in some inflammatory states.
Collapse
Affiliation(s)
- Kevin B. Walters
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison; Madison, WI 53706; USA
- Department of Pediatrics and Medical Microbiology and Immunology; University of Wisconsin-Madison; Madison, WI 53706; USA
| | - M. Ernest Dodd
- Department of Pediatrics and Medical Microbiology and Immunology; University of Wisconsin-Madison; Madison, WI 53706; USA
| | - Jonathan R. Mathias
- Department of Pediatrics and Medical Microbiology and Immunology; University of Wisconsin-Madison; Madison, WI 53706; USA
| | - Andrea J. Gallagher
- Department of Pediatrics and Medical Microbiology and Immunology; University of Wisconsin-Madison; Madison, WI 53706; USA
| | - David A. Bennin
- Department of Pediatrics and Medical Microbiology and Immunology; University of Wisconsin-Madison; Madison, WI 53706; USA
| | - Jennifer Rhodes
- Department of Pediatric Oncology; Dana-Farber Cancer Institute; Boston, MA 02115; USA
| | - John P. Kanki
- Department of Pediatric Oncology; Dana-Farber Cancer Institute; Boston, MA 02115; USA
| | - A. Thomas Look
- Department of Pediatric Oncology; Dana-Farber Cancer Institute; Boston, MA 02115; USA
| | - Yevgenya Grinblat
- Departments of Zoology and Anatomy, University of Wisconsin-Madison; Madison, WI 53706; USA
| | - Anna Huttenlocher
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison; Madison, WI 53706; USA
- Department of Pediatrics and Medical Microbiology and Immunology; University of Wisconsin-Madison; Madison, WI 53706; USA
| |
Collapse
|
147
|
Wollin M, Abele S, Bruns H, Weyand M, Kalden JR, Ensminger SM, Spriewald BM. Inhibition of TNF-alpha reduces transplant arteriosclerosis in a murine aortic transplant model. Transpl Int 2008; 22:342-9. [PMID: 19055619 DOI: 10.1111/j.1432-2277.2008.00802.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Experimental and clinical data provide evidence that TNF-alpha contributes to acute and chronic allograft rejection. In this study, we explored the effect of TNF-alpha blockade using the chimeric monoclonal antibody infliximab on the development of transplant arterisoclerosis in a fully mismatched aortic allograft model. Post-transplant treatment of CBA (H2(k)) recipients with 250 mug infliximab (cumulative dose 1.25 mg) reduced luminal occlusion of C57Bl/6 (H2(b)) aortic grafts on day 30 from 77 +/- 5% in untreated controls to 52 +/- 6%. Increasing the dose of anti-TNF-alpha antibody had no further beneficial effect. Treatment with human control immunoglobulin had no effect on intima proliferation. Under TNF-alpha blockade, ICAM-1 and PDGF mRNA expression within the grafts was strongly reduced, whereas iNOS expression was enhanced. The data show that TNF-alpha blockade using infliximab can reduce the development of transplant arteriosclerosis in fully mismatched murine aortic grafts.
Collapse
Affiliation(s)
- Martina Wollin
- Department for Internal Medicine 3 and Institute for Clinical Immunology, University Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
148
|
Roche JA, Lovering RM, Bloch RJ. Impaired recovery of dysferlin-null skeletal muscle after contraction-induced injury in vivo. Neuroreport 2008; 19:1579-84. [PMID: 18815587 PMCID: PMC2662728 DOI: 10.1097/wnr.0b013e328311ca35] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The protein, dysferlin, mediates sarcolemmal repair in vitro, implicating defective membrane repair in dysferlinopathies. To study the role of dysferlin in vivo, we assessed contractile function, sarcolemmal integrity, and myogenesis before and after injury from large-strain lengthening contractions in dysferlin-null and control mice. We report that dysferlin-null muscles produce higher contractile torque, and are equally susceptible to initial injury but recover from injury more slowly. Two weeks after injury, control muscles retain fluorescein dextran and do not show myogenesis. Dysferlin-null muscles do not retain fluorescein dextran, and show necrosis followed by myogenesis. Our data indicate that recovery of control muscles from injury primarily involves sarcolemmal repair whereas recovery of dysferlin-null muscles primarily involves myogenesis without repair and long-term survival of myofibers.
Collapse
Affiliation(s)
- Joseph A Roche
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
149
|
Grounds MD, Radley HG, Lynch GS, Nagaraju K, De Luca A. Towards developing standard operating procedures for pre-clinical testing in the mdx mouse model of Duchenne muscular dystrophy. Neurobiol Dis 2008; 31:1-19. [PMID: 18499465 PMCID: PMC2518169 DOI: 10.1016/j.nbd.2008.03.008] [Citation(s) in RCA: 266] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Revised: 03/20/2008] [Accepted: 03/24/2008] [Indexed: 11/19/2022] Open
Abstract
This review discusses various issues to consider when developing standard operating procedures for pre-clinical studies in the mdx mouse model of Duchenne muscular dystrophy (DMD). The review describes and evaluates a wide range of techniques used to measure parameters of muscle pathology in mdx mice and identifies some basic techniques that might comprise standardised approaches for evaluation. While the central aim is to provide a basis for the development of standardised procedures to evaluate efficacy of a drug or a therapeutic strategy, a further aim is to gain insight into pathophysiological mechanisms in order to identify other therapeutic targets. The desired outcome is to enable easier and more rigorous comparison of pre-clinical data from different laboratories around the world, in order to accelerate identification of the best pre-clinical therapies in the mdx mouse that will fast-track translation into effective clinical treatments for DMD.
Collapse
Affiliation(s)
- Miranda D Grounds
- School of Anatomy and Human Biology, the University of Western Australia, Perth, Western Australia, Australia.
| | | | | | | | | |
Collapse
|
150
|
Grounds MD, Radley HG, Gebski BL, Bogoyevitch MA, Shavlakadze T. IMPLICATIONS OF CROSS-TALK BETWEEN TUMOUR NECROSIS FACTOR AND INSULIN-LIKE GROWTH FACTOR-1 SIGNALLING IN SKELETAL MUSCLE. Clin Exp Pharmacol Physiol 2008; 35:846-51. [DOI: 10.1111/j.1440-1681.2007.04868.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|