Advertisement
Regular article Musculoskeletal pathology| Volume 179, ISSUE 5, P2464-2474, November 2011

Download started.

Ok

mdx5cv Mice Manifest More Severe Muscle Dysfunction and Diaphragm Force Deficits than Do mdx Mice

Open ArchivePublished:September 05, 2011DOI:https://doi.org/10.1016/j.ajpath.2011.07.009
      Duchenne muscular dystrophy (DMD) is characterized by progressive skeletal muscle dysfunction leading to premature death by the third decade of life. The mdx mouse, the most widely used animal model of DMD, has been extremely useful to study disease mechanisms and to screen new therapeutics. However, unlike patients with DMD, mdx mice have a very mild motor function deficit, posing significant limitations for its use as a platform to assess the impact of treatments on motor function. It has been suggested that an mdx variant, the mdx5cv mouse, might be more severely affected. Here, we compared the motor activity, histopathology, and individual muscle force measurements of mdx and mdx5cv mice. Our study revealed that mdx5cv mice showed more severe exercise-induced fatigue, Rotarod performance deficits, and gait anomalies than mdx mice and that these deficits began at a younger age. Muscle force studies showed more severe strength deficits in the diaphragm of mdx5cv mice compared to mdx mice, but similar force generation in the extensor digitorum longus. Muscle histology was similar between the two strains. Differences in genetic background (genetic modifiers) probably account for these functional differences between mdx strains. Overall, our findings indicate that the mdx and mdx5cv mouse models of DMD are not interchangeable and identify the mdx5cv mouse as a valuable platform for preclinical studies that require assessment of muscle function in live animals.
      Duchenne muscular dystrophy (DMD), the most prevalent muscular dystrophy, is an X-linked recessive disorder affecting 1 in 3500 male births. DMD is typically diagnosed around 3 years of age with a rapid progression of muscle weakness that results in wheelchair dependence by 12 years of age and death by the third decade of life.
      • Dubowitz V.
      Muscle Disorders in Childhood.
      Molecularly, DMD stems from mutations in the dystrophin gene that typically result in a lack of dystrophin protein expression in skeletal and cardiac muscles.
      • Hoffman E.P.
      • Brown Jr, R.H.
      • Kunkel L.M.
      Dystrophin: the protein product of the Duchenne muscular dystrophy locus.
      • Monaco A.P.
      • Neve R.L.
      • Colletti-Feener C.
      • Bertelson C.J.
      • Kurnit D.M.
      • Kunkel L.M.
      Isolation of candidate cDNAs for portions of the Duchenne muscular dystrophy gene.
      Loss of dystrophin compromises the muscle fiber membrane, leading to cycles of muscle fiber degeneration and regeneration, chronic inflammation, and accumulation of fibrotic tissue.
      • Matsumura K.
      • Ohlendieck K.
      • Ionasescu V.V.
      • Tome F.M.
      • Nonaka I.
      • Burghes A.H.
      • Mora M.
      • Kaplan J.C.
      • Fardeau M.
      • Campbell K.P.
      The role of the dystrophin-glycoprotein complex in the molecular pathogenesis of muscular dystrophies.
      • Engel A.G.
      • Ozawa E.
      Dystrophinopathies.
      Currently, there is no definitive treatment for DMD, but several gene-, cell-, and drug-based therapeutic approaches are being evaluated.
      • Zhou L.
      • Lu H.
      Targeting fibrosis in Duchenne muscular dystrophy.
      • Mitrpant C.
      • Fletcher S.
      • Wilton S.D.
      Personalised genetic intervention for Duchenne muscular dystrophy: antisense oligomers and exon skipping.
      • Nelson S.F.
      • Crosbie R.H.
      • Miceli M.C.
      • Spencer M.J.
      Emerging genetic therapies to treat Duchenne muscular dystrophy.
      • Muir L.A.
      • Chamberlain J.S.
      Emerging strategies for cell and gene therapy of the muscular dystrophies.
      • Peault B.
      • Rudnicki M.
      • Torrente Y.
      • Cossu G.
      • Tremblay J.P.
      • Partridge T.
      • Gussoni E.
      • Kunkel L.M.
      • Huard J.
      Stem and progenitor cells in skeletal muscle development, maintenance, and therapy.
      • Cossu G.
      • Sampaolesi M.
      New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials.
      Assessing and comparing the efficacy of these treatments require an easily available, well-characterized animal model with measurable and reproducible motor deficits, histologic pathology, and physiological alterations of muscle function.
      The mdx mouse is the most widely studied animal model of DMD and has been extensively used in preclinical studies over the past 20 years. It arose from a spontaneous nonsense mutation in exon 23 of the dystrophin gene in an inbred C57BL/10 background.
      • Bulfield G.
      • Siller W.G.
      • Wight P.A.
      • Moore K.J.
      X chromosome-linked muscular dystrophy (mdx) in the mouse.
      The mdx muscles lack dystrophin expression and show histopathological features similar to DMD.
      • Bulfield G.
      • Siller W.G.
      • Wight P.A.
      • Moore K.J.
      X chromosome-linked muscular dystrophy (mdx) in the mouse.
      • McGeachie J.K.
      • Grounds M.D.
      • Partridge T.A.
      • Morgan J.E.
      Age-related changes in replication of myogenic cells in mdx mice: quantitative autoradiographic studies.
      • Pastoret C.
      • Sebille A.
      mdx mice show progressive weakness and muscle deterioration with age.
      • Lefaucheur J.P.
      • Pastoret C.
      • Sebille A.
      Phenotype of dystrophinopathy in old mdx mice.
      However, contrary to DMD patients, mdx mice have a milder phenotype, with minimal fibrosis in all muscles with the exception of the diaphragm,
      • Stedman H.H.
      • Sweeney H.L.
      • Shrager J.B.
      • Maguire H.C.
      • Panettieri R.A.
      • Petrof B.
      • Narusawa M.
      • Leferovich J.M.
      • Sladky J.T.
      • Kelly A.M.
      The mdx mouse diaphragm reproduces the degenerative changes of Duchenne muscular dystrophy.
      a near normal life span,
      • Chamberlain J.S.
      • Metzger J.
      • Reyes M.
      • Townsend D.
      • Faulkner J.A.
      Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma.
      and only minimal motor deficits.
      • Bulfield G.
      • Siller W.G.
      • Wight P.A.
      • Moore K.J.
      X chromosome-linked muscular dystrophy (mdx) in the mouse.
      • Carnwath J.W.
      • Shotton D.M.
      Muscular dystrophy in the mdx mouse: histopathology of the soleus and extensor digitorum longus muscles.
      • Tanabe Y.
      • Esaki K.
      • Nomura T.
      Skeletal muscle pathology in X chromosome-linked muscular dystrophy (mdx) mouse.
      • Torres L.F.
      • Duchen L.W.
      The mutant mdx: inherited myopathy in the mouse Morphological studies of nerves, muscles and end-plates.
      • Dangain J.
      • Vrbova G.
      Muscle development in mdx mutant mice.
      • Muntoni F.
      • Mateddu A.
      • Marchei F.
      • Clerk A.
      • Serra G.
      Muscular weakness in the mdx mouse.
      In particular, treatment effect on overall motor function has been difficult to assess, posing a challenge for preclinical studies.
      • Grounds M.D.
      • Radley H.G.
      • Lynch G.S.
      • Nagaraju K.
      • De Luca A.
      Towards developing standard operating procedures for pre-clinical testing in the mdx mouse model of Duchenne muscular dystrophy.
      • Spurney C.F.
      • Gordish-Dressman H.
      • Guerron A.D.
      • Sali A.
      • Pandey G.S.
      • Rawat R.
      • Van Der Meulen J.H.
      • Cha H.J.
      • Pistilli E.E.
      • Partridge T.A.
      • Hoffman E.P.
      • Nagaraju K.
      Preclinical drug trials in the mdx mouse: assessment of reliable and sensitive outcome measures.
      As a result, mdx variants were generated with the hope of obtaining a more severe phenotype.
      • Chapman V.M.
      • Miller D.R.
      • Armstrong D.
      • Caskey C.T.
      Recovery of Induced Mutations for X Chromosome-Linked Muscular Dystrophy in Mice.
      • Araki E.
      • Nakamura K.
      • Nakao K.
      • Kameya S.
      • Kobayashi O.
      • Nonaka I.
      • Kobayashi T.
      • Katsuki M.
      Targeted disruption of exon 52 in the mouse dystrophin gene induced muscle degeneration similar to that observed in Duchenne muscular dystrophy.
      • Fukada S.-i.
      • Morikawa D.
      • Yamamoto Y.
      • Yoshida T.
      • Sumie N.
      • Yamaguchi M.
      • Ito T.
      • Miyagoe-Suzuki Y.
      • Takeda Si
      • Tsujikawa K.
      • Yamamoto H.
      Genetic background affects properties of satellite cells and mdx phenotypes.
      However, these alternative DMD mouse models had histopathological features and mild functional impairment similar to the mdx mouse. Among these, mdx5cv mice harbor a mutation affecting exon 10 that selectively disrupts expression of full-length dystrophin such as in mdx mice.
      • Im W.B.
      • Phelps S.F.
      • Copen E.H.
      • Adams E.G.
      • Slightom J.L.
      • Chamberlain J.S.
      Differential expression of dystrophin isoforms in strains of mdx mice with different mutations.
      However, mdx5cv mice have recently been preferred to mdx mice because of their very low level of dystrophin-expressing revertant fibers,
      • Danko I.
      • Chapman V.
      • Wolff J.A.
      The frequency of revertants in mdx mouse genetic models for Duchenne muscular dystrophy.
      allowing a more accurate quantification of the efficiency of gene and cell therapy in restoring dystrophin expression. In addition, mdx5cv mice are on the C57BL/6 background that allows rapid transfer of the dystrophin mutation onto transgenic and knock-out mice to dissect molecular aspects of disease.
      Although mdx and mdx5cv mice have been generally regarded as similar, a recent study found significant differences in gene expression profiles in all muscle groups examined, and a qualitative histologic examination suggested a more severe pathology in mdx5cv mice.
      • Haslett J.N.
      • Kang P.B.
      • Han M.
      • Kho A.T.
      • Sanoudou D.
      • Volinski J.M.
      • Beggs A.H.
      • Kohane I.S.
      • Kunkel L.M.
      The influence of muscle type and dystrophin deficiency on murine expression profiles.
      We therefore sought to determine whether mdx5cv mice have more severe motor function deficits that render them distinct from mdx mice and better suited to study the effect of treatments on motor function and endurance.

      Materials and Methods

      Experimental Animals

      All experimental mice were bred in-house and kept under similar conditions. Original C57BL/6J and C57BL/10SnJ breeders were obtained from The Jackson Laboratory (Bar Harbor, ME). C57BL/10ScSn-Dmdmdx (referred to as mdx) and BL6Ros.Cg-Dmdmdx5cv (referred to as mdx5cv) breeders were kind gifts from Dr. Arthur Burghes (Ohio State University, Columbus, OH) and Dr. Louis Kunkel (Harvard Medical School, Boston, MA), respectively. Breeders were fed a high-fat diet (Teklad Global 19% protein rodent diet, no. 2019; Harlan, Indianapolis, IN), whereas weaned mice were switched to a low-fat diet (Teklad Global 18% protein rodent diet, no. 2018; Harlan). Animal breeding and experimental procedures followed approved protocols by the Institutional Animal Care and Use Committee. Unless otherwise stated, male mice were used.

      Body and Individual Muscle Weights

      Body weights were measured once a week from multiple litters. For individual muscle weights, the quadriceps, gastrocnemius, tibialis anterior, and triceps muscles from left and right limbs as well as the diaphragm were carefully dissected. Muscles were cleaned from fat and tendons before weighing. Weights of the left and right muscles for each individual mouse were averaged and then normalized to the tibia length (mg/cm).

      Tibia Length

      The tibia, knee, and ankle joints from the left hind limb were cleaned from tissue. Hind limbs were photographed, and tibia length from the knee to the ankle joint was measured with ImageJ software version 1.39u (NIH, Bethesda, MD).

      Rotarod Assay

      Ten to 15 mice per group (C57BL/6, C57BL/10, mdx and mdx5cv) were tested on a Rotarod apparatus (Columbus Instruments, Columbus, OH) once a week in the morning, following a previously described protocol.
      • Payne E.T.
      • Yasuda N.
      • Bourgeois J.M.
      • Devries M.C.
      • Rodriguez M.C.
      • Yousuf J.
      • Tarnopolsky M.A.
      Nutritional therapy improves function and complements corticosteroid intervention in mdx mice.
      Briefly, mice were placed on the stationary rod that was gradually brought to a constant speed of 20 rpm. At this point, the latency to fall was recorded with a maximum time on the rod of 5 minutes (300 seconds). Results from three trials (with a 1-minute rest between trials) per mouse per session were recorded and averaged. Mice were acclimated 2 weeks before first data collection.

      Exercise-Induced Fatigue Assay

      A previously described protocol
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      was followed with the following modifications. Activity measurements were taken on a Photobeam Activity System (San Diego Instruments, San Diego, CA) under dim light in the early morning before and after a single bout of treadmill exercise. Activity was recorded for 1 hour at 5-minutes interval. The DMD_M.2.1.002 TREAT-NMD standard operating procedure (SOP; Experimental Protocols for DMD Animal Models, http://www.treat-nmd.eu/research/preclinical/dmd-sops, last accessed June 30, 2011) was followed, and the first 5 minutes of recorded activity were discarded from analysis. Treadmill (Columbus Instruments) settings were 15° decline, 10 m/min speed, with a 10-minute run. For each individual mouse, post-exercise values were normalized to pre-exercise levels as follows. Ambulation, rearing, and distance traveled are expressed as the percentage of change in post-exercise levels compared with pre-exercise levels: [(Activity after exercise/Activity before exercise) × 100] – 100. Pre-exercise rest time values were subtracted from post-exercise values.

      Grip Test

      The M.2.2_001TREAT-NMD SOP was followed. Five grip strength measurements for forelimbs and hind limbs were taken for 5 to 7 mice per group on a strength meter (Columbus Instruments) and averaged. Force measurements (g) were normalized to body weight (g).

      Footprint Test

      The footprint test was performed as previously described.
      • Carter R.J.
      • Lione L.A.
      • Humby T.
      • Mangiarini L.
      • Mahal A.
      • Bates G.P.
      • Dunnett S.B.
      • Morton A.J.
      Characterization of progressive motor deficits in mice transgenic for the human Huntington's disease mutation.
      Briefly, hind paws and forepaws of mice were painted blue and red, respectively. Mice were allowed to walk along a 50-cm long and 10-cm wide corridor lined with paper to record their footprints. Three parameters were measured (see Supplemental Figure S1 at http://ajp.amjpathol.org) on tracings where a consistent straight gait pattern was achieved: hind paw base width (distance between left and right hind paw footprints), separation (distance between the center of the front and hind footprints of the same side), and stride length. For each mouse, measurements from two to three sets of footprints per trial were averaged. Six to 10 male mice per group were analyzed at 8 weeks, 12 weeks, and 1 year of age.

      Evans Blue Dye Injections and Quantifications

      Evans Blue dye (MP Biomedicals, Inc., Irvine, CA) was administered intraperitoneally (5 μL/g of body weight). For visualization on tissue sections, Evans Blue dye was administered 16 to 24 hours before sacrifice.
      • Hamer P.W.
      • McGeachie J.M.
      • Davies M.J.
      • Grounds M.D.
      Evans Blue Dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability.
      For spectrophotometric quantifications, mice were injected 40 hours before sacrifice, and tissues were analyzed as previously described.
      • Heydemann A.
      • Ceco E.
      • Lim J.E.
      • Hadhazy M.
      • Ryder P.
      • Moran J.L.
      • Beier D.R.
      • Palmer A.A.
      • McNally E.M.
      Latent TGF-beta-binding protein 4 modifies muscular dystrophy in mice.
      Optical density values at 260 nm (SpectraMax M2; Molecular Devices, Sunnyvale, CA) were normalized to the individual muscle weight (mg).

      Immunostaining for CD45 and Collagen III

      Immunostaining of CD45 and collagen III were done as previously described
      • Zhou L.
      • Rafael-Fortney J.A.
      • Huang P.
      • Zhao X.S.
      • Cheng G.
      • Zhou X.
      • Kaminski H.J.
      • Liu L.
      • Ransohoff R.M.
      Haploinsufficiency of utrophin gene worsens skeletal muscle inflammation and fibrosis in mdx mice.
      on 8-μm frozen sections with the following primary antibodies: rat anti-mouse CD45 (1:200; AbD Serotec, Raleigh, NC) and collagen III (1:20; Southern Biotech, Birmingham, AL). For CD45 staining, antibody binding was visualized with DAB substrate (Vector Laboratories, Burlington, ON, Canada) and viewed under a light microscope. For collagen III staining, antibody signal was visualized with a fluorescein-conjugated secondary antibody and viewed under a fluorescence microscope.

      Quantification of Inflammation and Collagen Deposition from Tissue Sections

      Quantification was done as previously described.
      • Huang P.
      • Zhao X.S.
      • Fields M.
      • Ransohoff R.M.
      • Zhou L.
      Imatinib attenuates skeletal muscle dystrophy in mdx mice.
      The percentage of areas immunoreactive for CD45 and collagen III were quantified on five sections from each specimen separated by at least 50 μm. Five nonoverlapping areas of each section were digitally captured with the use of an Olympus microscope (Olympus, Center Valley, PA) equipped with a high-resolution digital camera. The immunoreactive area was determined for each marker after manual outlining with the use of ImageJ software (NIH) and was expressed as a percentage of the visualized section area. The average from five sections was calculated for statistical comparisons (mean ± SD).

      Hydroxyproline Assay

      Hydroxyproline concentration of muscle samples from 12-week-old and 1-year-old mdx and mdx5cv mice (5 mice per group) was analyzed as previously described.
      • Reddy G.K.
      • Enwemeka C.S.
      A simplified method for the analysis of hydroxyproline in biological tissues.

      Force Measurements in Diaphragm and EDL Muscles

      The extensor digitorum longus (EDL) and diaphragm muscle strips were isolated immediately after euthanasia while the heart was still beating and bathed in oxygenated Krebs-Henseleit solution [95% O2/5% CO2 (pH 7.4), 118 mmol/L NaCl, 25 mmol/L NaHCO3, 5 mmol/L KCl, 1 mmol/L KH2PO4, 2 mmol/L CaCl2, 1 mmol/L MgCl2, 5 mmol/L glucose] in a circulating bath. Diaphragm contractile strength measurements were done as previously described.
      • Acharyya S.
      • Villalta S.A.
      • Bakkar N.
      • Bupha-Intr T.
      • Janssen P.M.
      • Carathers M.
      • Li Z.W.
      • Beg A.A.
      • Ghosh S.
      • Sahenk Z.
      • Weinstein M.
      • Gardner K.L.
      • Rafael-Fortney J.A.
      • Karin M.
      • Tidball J.G.
      • Baldwin A.S.
      • Guttridge D.C.
      Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy.
      • Chandrasekharan K.
      • Yoon J.H.
      • Xu Y.
      • deVries S.
      • Camboni M.
      • Janssen P.M.
      • Varki A.
      • Martin P.T.
      A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy.
      • Peterson J.M.
      • Kline W.
      • Canan B.D.
      • Ricca D.J.
      • Kaspar B.
      • Delfin D.A.
      • Dirienzo K.
      • Clemens P.R.
      • Robbins P.D.
      • Baldwin A.S.
      • Flood P.
      • Kaumaya P.
      • Frietas M.
      • Kornegay J.N.
      • Mendell J.R.
      • Rafael-Fortney J.A.
      • Guttridge D.C.
      • Janssen P.M.
      Peptide-based inhibition of NF-kappaB rescues diaphragm muscle contractile dysfunction in a murine model of Duchenne muscular dystrophy.
      This protocol deviated from the M.1.2_002 TREAT-NMD SOP mainly in that strength was assessed at 37°C. This temperature was chosen because it is physiologically more relevant to the in vivo function of diaphragm muscle, and reliable measurements can be performed as shown by us
      • Acharyya S.
      • Villalta S.A.
      • Bakkar N.
      • Bupha-Intr T.
      • Janssen P.M.
      • Carathers M.
      • Li Z.W.
      • Beg A.A.
      • Ghosh S.
      • Sahenk Z.
      • Weinstein M.
      • Gardner K.L.
      • Rafael-Fortney J.A.
      • Karin M.
      • Tidball J.G.
      • Baldwin A.S.
      • Guttridge D.C.
      Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy.
      • Chandrasekharan K.
      • Yoon J.H.
      • Xu Y.
      • deVries S.
      • Camboni M.
      • Janssen P.M.
      • Varki A.
      • Martin P.T.
      A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy.
      and others.
      • Rucker M.
      • Fraites Jr, T.J.
      • Porvasnik S.L.
      • Lewis M.A.
      • Zolotukhin I.
      • Cloutier D.A.
      • Byrne B.J.
      Rescue of enzyme deficiency in embryonic diaphragm in a mouse model of metabolic myopathy: pompe disease.
      • Diaz P.T.
      • Brownstein E.
      • Clanton T.L.
      Effects of N-acetylcysteine on in vitro diaphragm function are temperature dependent.
      • Prezant D.J.
      • Richner B.
      • Valentine D.E.
      • Aldrich T.K.
      • Fishman C.L.
      • Nagashima H.
      • Chaudhry I.
      • Cahill J.
      Temperature dependence of rat diaphragm muscle contractility and fatigue.
      Briefly, small linear strips were dissected from the diaphragm and connected to a linear micromanipulator and a fixed hook. Electrical stimulation was achieved with parallel platinum-iridium electrodes. First, twitch contractions were used, and the muscle was stretched until twitches produced maximal force. Thereafter, a paired-pulse protocol was used, and various frequencies of pulse intervals were used, with 10 minutes of rest between each stimulation. At 150 Hz, a smooth tetanus was obtained with the use of this protocol. After a final rest period, fatigue was assessed by sending a 100-Hz (500 milliseconds) impulse every second for 1 minute. After force measurements were taken, tissues were only lightly blotted before weighing to allow use of the same muscle for histologic analysis. This resulted in a 30% to 40% overestimation of muscle weight. As a result, when normalized isometric force measurements (mN/mm2) per unit of cross-sectional area is based on total weight (using muscle density of 1.06 g/cm3), these values are generally 30% to 40% lower than when calculated after complete tissue blotting. Because we did record all 3 muscle dimensions for diaphragm strips (using a stereo dissection microscope at ×40), specific force was calculated by dividing total active force by the central cross-sectional area.
      EDL force measurements were taken at 30°C as described previously.
      • Chandrasekharan K.
      • Yoon J.H.
      • Xu Y.
      • deVries S.
      • Camboni M.
      • Janssen P.M.
      • Varki A.
      • Martin P.T.
      A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy.
      • Rodino-Klapac L.R.
      • Janssen P.M.
      • Montgomery C.L.
      • Coley B.D.
      • Chicoine L.G.
      • Clark K.R.
      • Mendell J.R.
      A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy.
      • Martin P.T.
      • Xu R.
      • Rodino-Klapac L.R.
      • Oglesbay E.
      • Camboni M.
      • Montgomery C.L.
      • Shontz K.
      • Chicoine L.
      • Clark K.R.
      • Sahenk Z.
      • Mendell J.R.
      • Janssen P.M.
      Overexpression of Galgt2 in skeletal muscle prevents injury resulting from eccentric contractions in both mdx and wild type mice.
      Briefly, one end of the EDL muscle was tied to a force transducer and the other to a high-speed linear servo-controlled motor. At a resting tension of 1 × g, stimulation was delivered by two parallel platinum-iridium electrodes. Muscles were adjusted to optimum length, defined as the length for maximal twitch when subjected to an isometric tetanus of 150 Hz, for 500 milliseconds. After a 10-minute rest period, muscles were subjected to an eccentric contraction protocol that consisted of a series of 10 isometric 700-millisecond tetani, at 2-minute intervals, with a 5% lengthening of the muscles (0.5 fiber length per second for duration of 200 milliseconds) when maximal force has developed at 500 milliseconds. When the tetanus ended (at t = 700 milliseconds), the muscle was brought back to initial length (at the same speed as the stretch), allowing for full relaxation to the initial length. For EDL muscle, specific force was calculated with the recorded weights and is thus ∼30% to 40% lower because of the mild blotting used to preserve histology. All groups were treated identically.

      AAV8.MCK.microdystrophin Vector Production and Administration to Mice

      High-titer recombinant AAV8 vectors were produced by a modified cross-packaging approach.
      • Rabinowitz J.E.
      • Rolling F.
      • Li C.
      • Conrath H.
      • Xiao W.
      • Xiao X.
      • Samulski R.J.
      Cross-packaging of a single adeno-associated virus (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity.
      The murine microdystrophin construct R4-R23/Δ71-72
      • Rodino-Klapac L.R.
      • Janssen P.M.
      • Montgomery C.L.
      • Coley B.D.
      • Chicoine L.G.
      • Clark K.R.
      • Mendell J.R.
      A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy.
      was placed under the control of a truncated MCK (muscle creatine kinase) promoter/enhancer to drive muscle-specific gene expression (AAV8.MCK.microdystrophin).
      • Hauser M.A.
      • Amalfitano A.
      • Kumar-Singh R.
      • Hauschka S.D.
      • Chamberlain J.S.
      Improved adenoviral vectors for gene therapy of Duchenne muscular dystrophy.
      Five neonatal mdx5cv mice received an intraperitoneal injection of 3.7 × 1011 AAV8.MCK.microdystrophin vector genomes in a total volume of 100 μL.

      Dystrophin Immunostaining

      Diaphragm, quadriceps, gastrocnemius, tibialis anterior, and triceps muscles were embedded in 7% gum tragacanth and flash frozen in liquid nitrogen. Unfixed 12-μm cryosections were processed for immunohistochemistry as previously described
      • Rodino-Klapac L.R.
      • Janssen P.M.
      • Montgomery C.L.
      • Coley B.D.
      • Chicoine L.G.
      • Clark K.R.
      • Mendell J.R.
      A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy.
      with the use of the anti-dystrophin Manex1a and Manex1011b antibodies (Developmental Studies Hybridoma Bank, Iowa City, IA; 1:50) that also recognize microdystrophin. Fluorescence images were captured under a ×20 objective with a Hamamatsu ORCA-ER digital camera mounted on an Olympus BX61 microscope. Montage images (Slidebook 4.2; Intelligent Imaging Innovations, Denver, CO) of the entire tissue section were used to manually quantify the number of total and microdystrophin-positive myofibers. Values were used to calculate the percentage of microdystrophin-positive fibers.

      Western Blot Analysis

      Five, 50-μm diaphragm tissue sections were homogenized in ice-cold protein lysis buffer [125 mmol/L Tris-HCl, 4% SDS, 4M urea, 5% 2-Mercaptoethanol, protease inhibitor cocktail (Roche, Indianapolis, IN)] with the use of a pellet pestle motor (Kimble Chase, Vineland, NJ). Protein lysates were centrifuged at 14,000 × g for 15 minutes at 4°C to remove insoluble material. Protein concentration was determined on a Nanodrop 1000 (Thermo Scientific, Waltham, MA) before adding 4× SDS-PAGE reducing sample buffer. Proteins were resolved on a 4% to 12% gradient SDS Bis-Tris gel (Invitrogen, Carlsbad, CA) and transferred to a nitrocellulose membrane that was cut at the 60-kDa molecular weight mark. The top membrane (molecular weight >60 kDa) was incubated with the Manex1011b antibody (1:1000; Developmental Studies Hybridoma Bank) to detect dystrophin and microdystrophin, whereas the bottom was used for protein loading control with the use of an anti-actin antibody (1:500; clone 20 to 33; Sigma-Aldrich, St. Louis, MO). After incubation with horseradish peroxidase-conjugated secondary antibodies (Jackson ImmunoResearch Laboratories Inc., West Grove, PA), signal was detected by chemiluminescence (Thermo Scientific) on X-ray film (Research Products International, Mt. Prospect, IL).

      Densitometric Analysis

      Densitometric analysis was performed with the ImageJ software (NIH) on scanned X-ray films taken at nonsaturating exposures. Microdystrophin and dystrophin band intensities were normalized to actin for each sample. Percentages of microdystrophin levels were calculated relative to dystrophin in wild-type mice.

      Quantification of Diaphragm Myofibers with Centrally Located Nuclei

      Two 12-μm diaphragm sections were stained with hematoxylin and eosin. Bright-field pictures were taken with a 10× objective and a color Olympus DP71 camera mounted on a Zeiss Axioskop microscope (Zeiss, Thornwood, NY). For each section, two random nonoverlapping fields were photographed, and the numbers of total myofibers and of myofibers with a centrally located nucleus were counted and used to calculate the following ratio per mouse: total number of myofibers with central nuclei/the total number of myofibers counted.

      Statistical Analyses

      For comparisons between two groups we used a two-tailed unpaired Student's t test (significance set at P < 0.05). For multiple group comparisons, a two-way analysis of variance followed by the Bonferroni test for pair-wise comparisons was used. For experiments with repeated measures, a repeated-measure analysis of variance was performed to detect significant differences between groups.

      Results

      Differences in Body and Muscle Weights between mdx5cv and mdx Mice

      We first determined whether significant weight or growth differences were present between mdx and mdx5cv mice because they might affect parameters measured in motor function tests. In agreement with previous reports,
      • Pastoret C.
      • Sebille A.
      mdx mice show progressive weakness and muscle deterioration with age.
      • Cohn R.D.
      • Liang H.Y.
      • Shetty R.
      • Abraham T.
      • Wagner K.R.
      Myostatin does not regulate cardiac hypertrophy or fibrosis.
      • Hayes A.
      • Williams D.A.
      Contractile function and low-intensity exercise effects of old dystrophic (mdx) mice.
      we found that mdx mice had a significantly lower body weight than wild-type mice between 8 and 20 weeks of age (Figure 1A). mdx5cv mice also had a significantly lower body weight compared with wild-type controls but also compared with mdx mice at most ages tested. This difference could not be attributed to intrinsic strain differences because C57BL/6 and C57BL/10 wild-type mice had similar body weights.
      Figure thumbnail gr1
      Figure 1Comparison of body and muscle weight among wild-type, mdx, and mdx5cv mice. A: Body weight of wild-type and dystrophic mice between 8 and 20 weeks of age. mdx5cv mice have a significantly lower body weight than wild-type mice at all ages studied (P < 0.05). Significant differences between mdx and mdx5cv mice are indicated. *P < 0.05, **P < 0.01. Values are means ± SEs (n = 5 to 25 mice per group). B: Individual muscle weight normalized to tibia length for 42-week-old male mice. Data are means ± SDs (n = 4). QD, quadriceps; TA, tibialis anterior; GS, gastrocnemius; TC, triceps; DIA, diaphragm. No statistically significant differences were found.
      Because we observed more abdominal fat accumulation in wild-type mice than in either dystrophic mouse strain, we measured tibia length, an alternative measure of growth that is linked to muscle mass but independent of fat.
      • Hooper A.C.
      Bone length and muscle weight in mice subjected to genetic selection for the relative length of the tibia and radius.
      Tibia length showed no difference between wild-type and dystrophic mice or between mdx and mdx5cv mice (see Supplemental Figure S2, A and B, at http://ajp.amjpathol.org). Furthermore, measurements of body length (base of the neck to the base of the tail) taken after sacrifice in 1-year-old mice were similar between all strains (data not shown). These results suggest that weight differences between dystrophic and wild-type mice as well as between mdx and mdx5cv mice do not reflect differences in growth or overall mouse size but may reflect differences in body fat. To further test for differences between mdx and mdx5cv mice in muscles involved in motor function, we compared the weight of freshly isolated limb and diaphragm muscles from male dystrophic mice and found no significant differences (Figure 1B). Therefore, mdx and mdx5cv mice are similar in size and individual muscle weight, allowing for a direct comparison of their motor function.

      Differences in Rotarod Performance, Fatigue, and Gait between mdx5cv and mdx Mice

      Next, we evaluated three modalities of muscle function in male mice: Rotarod performance, exercise-induced fatigue, and gait. For all tests performed, we found no significant differences between wild-type C57BL/6 and C57BL/10 mice (Figure 2A; see also Supplemental Figures S3 and S4 at http://ajp.amjpathol.org). For simplicity, we are showing C57BL/6 data to highlight the severity of motor deficits in dystrophic strains relative to wild-type performance. Rotarod performance was assayed with a protocol that challenges both motor coordination and endurance.
      • Payne E.T.
      • Yasuda N.
      • Bourgeois J.M.
      • Devries M.C.
      • Rodriguez M.C.
      • Yousuf J.
      • Tarnopolsky M.A.
      Nutritional therapy improves function and complements corticosteroid intervention in mdx mice.
      The performance of mdx mice was not significantly different from either C57BL/10 or C57BL/6 wild-type mice, although it appeared mildly affected after 10 weeks of age (Figure 2A). In stark contrast, mdx5cv mice showed a significant motor deficit at all ages compared with both wild-type and mdx mice which was particularly pronounced between 6 and 10 weeks. Therefore, the Rotarod assay showed a deficit in motor coordination and possibly endurance that is only present in mdx5cv mice.
      Figure thumbnail gr2
      Figure 2Rotarod, exercise-induced fatigue, and gait analysis in dystrophic and wild-type mice. A: Rotarod performance. mdx5cv mice have a significant deficit in Rotarod performance compared with either wild-type or mdx mice (*P < 0.05, **P < 0.01, and ***P < 0.005). No significant difference was found between mdx and wild-type mice. Data are mean ± SE (n = 5 to 15 mice). B–D: Decrease in rearing events (B), ambulation (C), and distance traveled (D) after exercise compared with pre-exercise levels. Data are mean ± SD expressed as percentage of change in activity (n = 5 to 12 mice). E: Increase in rest time after exercise. Data are mean ± SD (n = 5 to 12 mice). Values are normalized to pre-exercise levels. F–H: Measurements of hind paw base width (F), hind paw to forepaw separation (G), and stride length (H) in wild-type C56BL/6, mdx, and mdx5cv mice. Data represent mean ± SE. B–H: Significant differences: *P < 0.05, **P < 0.01, and ***P < 0.005.
      To evaluate separately endurance and motor coordination, we tested exercise-induced fatigue and gait, respectively. In preliminary tests, we observed that contrary to mdx mice, mdx5cv mice were unable to sustain 30 minutes of downhill treadmill running and showed extreme fatigue after 15 minutes. To quantify this difference in exercise-induced fatigue, we compared open field activity of wild-type, mdx5cv, and mdx mice at 8 weeks, 12 weeks, and 1 year of age before and after a single 10-minute bout of treadmill running. As previously reported,
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      we found no significant differences in baseline levels of ambulation (horizontal activity), rearing events (vertical activity), distance traveled, or resting time between wild-type and dystrophic mice (data not shown). To account for interindividual variability, activity values measured after exercise were normalized to pre-exercise levels for each individual mouse (see Materials and Methods). Overall, exercise decreased ambulation, rearing activity, and the distance traveled, and it increased rest time in both wild-type and dystrophic mice, with more pronounced effects in dystrophic mice (Figure 2, B–E). In agreement with previous findings,
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      only rearing events were significantly decreased after exercise in young (8 weeks) mdx mice compared with wild-type mice (Figure 2B). By 12 weeks of age, mdx mice also showed a significant decrease in ambulation and increased rest time (Figure 2, C and E), whereas the distance traveled was significantly decreased only at 1 year of age (Figure 2D). Thus, mdx mice became more sensitive to exercise-induced fatigue with age. By contrast, mdx5cv mice showed maximum decreases (>90% drop in activity after exercise) in both ambulation and rearing events, starting as early as 8 weeks of age (Figure 2, B and C). By 12 weeks of age, mdx5cv mice showed a significant decrease in distance traveled compared with both wild-type and mdx mice. Interestingly, mdx5cv mice had a surprisingly uniform response to exercise-induced fatigue, whereas mdx mice showed a high level of interindividual variability. The coefficient of variation at 12 weeks was 2.5% for mdx5cv mice (n = 8) compared with 26.2% for mdx mice (n = 7) for ambulation, and 1.0% versus 17.1%, respectively, for rearing events. These results indicate that mdx5cv mice are more sensitive than mdx mice to exercise-induced fatigue, are affected at an earlier age, and show minimal interindividual variability in this assay.
      Motor coordination in dystrophic mice was further evaluated with gait analysis. Visual comparison of gait tracings between wild-type, mdx, and mdx5cv mice indicated that dystrophic mice, and in particular mdx5cv mice, had a shorter stride and that their hind paw did not always land on the same position as the front paw as is normal for mice (see Supplemental Figure S1A at http://ajp.amjpathol.org). Gait abnormalities were confirmed by measurements of hind paw base width (Figure 2F), front-hind paw separation (Figure 2G), and stride length (Figure 2H). Both mdx and mdx5cv mice had a significantly larger hind paw base width than wild-type mice, possibly indicative of muscle weakness. However, measurements of front-hind paw separation and especially stride length showed significant gait abnormalities that were more severe in mdx5cv mice than in wild-type and mdx mice. This could not be attributed to a smaller body size because tibia length measurements and postmortem examinations showed no differences between strains. Of note, 1-year-old wild-type mice show increased base width and separation, becoming indistinguishable from dystrophic mice. Therefore, the gait test is sensitive to age. Taken together, the above tests all indicate deficits in motor coordination, endurance, and gait that are present earlier, are more severe, and progress more rapidly in mdx5cv than in mdx mice.

      Differences in the Contractile Properties of the Diaphragm between mdx and mdx5cv Mice

      We next evaluated whether differences in the severity of motor function deficits between mdx and mdx5cv mice could stem from differences in limb and/or diaphragm muscle strength. Whole-limb muscle strength was evaluated with the grip test. In agreement with previous studies,
      • Golumbek P.T.
      • Keeling R.M.
      • Connolly A.M.
      Strength and corticosteroid responsiveness of mdx mice is unchanged by RAG2 gene knockout.
      • Wang B.
      • Li J.
      • Fu F.H.
      • Xiao X.
      Systemic human minidystrophin gene transfer improves functions and life span of dystrophin and dystrophin/utrophin-deficient mice.
      both mdx and mdx5cv mice showed a significant deficit in grip strength compared with wild-type mice for both hind limbs and forelimbs (Figure 3A; see also Supplemental Figure S5A at http://ajp.amjpathol.org). However, no difference was found between mdx and mdx5cv mice. Similarly, ex vivo measurements of tetanic force and resistance to eccentric contraction-induced damage in the EDL showed no significant differences between mdx and mdx5cv mice (see Supplemental Figure S5, B and C, at http://ajp.amjpathol.org). Therefore, limb muscles of mdx5cv mice do not appear to be weaker or more sensitive to eccentric contraction-induced damage than mdx mice.
      Figure thumbnail gr3
      Figure 3Limb grip strength and diaphragm force measurements in wild-type, mdx, and mdx5cv mice. A: Grip strength assay of hind limbs and forelimbs of 42-week-old mice. No significant difference was found between mdx and mdx5cv mice. Grip strength was normalized to body weight. Data are mean ± SD (n = 5 to 6 mice). Significant differences from wild-type mice: *P < 0.05, **P < 0.01. B: Tetanic force of diaphragm muscle. mdx and mdx5cv mice showed significantly lower force generation (smooth tetanus at 150 Hz) than strain-matched wild-type mice. In addition, a significantly lower tetanic force was generated by the diaphragm of mdx5cv than mdx mice. Significant differences between groups: *P < 0.05, ***P < 0.005. C: Diaphragm muscle response to the repetitive isometric contraction protocol. The diaphragm of mdx5cv mice showed enhanced susceptibility to fatigue compared with wild-type and mdx mice (*P < 0.05). B and C: Data are mean ± SE (n = 10/group).
      By contrast, the diaphragm of mdx5cv mice showed a significantly lower tetanic force (Figure 3B) and enhanced susceptibility to fatigue during 60 repetitive tetani (Figure 3C) compared with mdx mice. These differences could not be attributed to baseline differences between the C57BL/6 and C57BL/10 strains (Figure 3, B and C). Diaphragm force and fatigue measurements in both mdx and mdx5cv mice were significantly lower than in wild-type mice. Therefore, mdx5cv mice have a more severe deficit in force generation and resistance to fatigue in the diaphragm and their limb muscle strength is equivalent to mdx mice.

      Histopathological Parameters in the Diaphragm Are Similar between mdx and mdx5cv Mice

      To determine whether the diaphragm of mdx5cv mice had more pronounced histopathology, we quantified and compared connective tissue deposition, immune cell infiltration, and muscle fiber membrane permeability in the diaphragm muscles of mdx5cv and mdx mice. We further extended our analysis to limb muscles to complement our muscle strength analysis. We studied mice at 12 weeks when significant differences in motor performance were present between mdx5cv and mdx mice. We also compared old (45 to 53 weeks of age) mdx5cv and mdx mice when pathology is most evident.
      Collagen deposition, an indicator of fibrosis, was quantified in the diaphragm and quadriceps muscles by measuring hydroxyproline content and by immunohistochemistry. For both mdx5cv and mdx mice, the diaphragm showed increased collagen deposition between 12 weeks and 1 year of age and higher hydroxyproline content than the quadriceps (Figure 4, A–D). However, no significant differences were found between mdx and mdx5cv mice in either tissue at either age, suggesting comparable levels of fibrosis. Similarly, quantification of global immune cell infiltration (lymphocytes and macrophages) did not show significant differences between mdx and mdx5cv mice for either diaphragm or quadriceps muscles (Figure 5, A and B). Finally, quantification of Evans Blue dye incorporation, a measure of myofiber damage,
      • Hamer P.W.
      • McGeachie J.M.
      • Davies M.J.
      • Grounds M.D.
      Evans Blue Dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability.
      • Zeman R.J.
      • Peng H.
      • Danon M.J.
      • Etlinger J.D.
      Clenbuterol reduces degeneration of exercised or aged dystrophic (mdx) muscle.
      • Wooddell C.I.
      • Zhang G.
      • Griffin J.B.
      • Hegge J.O.
      • Huss T.
      • Wolff J.A.
      Use of Evans blue dye to compare limb muscles in exercised young and old mdx mice.
      • Straub V.
      • Rafael J.A.
      • Chamberlain J.S.
      • Campbell K.P.
      Animal models for muscular dystrophy show different patterns of sarcolemmal disruption.
      also failed to show differences between mdx and mdx5cv mice in the diaphragm as well as quadriceps, tibialis anterior, gastrocnemius, and triceps muscles at either 12 weeks or 1 year of age (Figure 5, C and D; data not shown). Similar to fibrosis, only the diaphragm showed a significant increase in Evans Blue dye incorporation in older mice (Figure 5C). These results suggest that mdx and mdx5cv mice are similar for the histopathological parameters we evaluated here. We found no evidence of more severe histopathology in the diaphragm of mdx5cv mice that could account for a more severe force deficit compared with mdx mice.
      Figure thumbnail gr4
      Figure 4Quantification of fibrosis in the quadriceps and diaphragm muscles of wild-type, mdx, and mdx5cv mice. A: Hydroxyproline content in diaphragm muscles of 12-week-old and 1-year-old mdx and mdx5cv mice. No statistically significant differences were found. B: Hydroxyproline content in quadriceps muscles of 12-week-old and 1-year-old mdx and mdx5cv mice. No statistically significant differences were found. Note difference in scale between panels A and B. C: Representative micrographs of diaphragm muscle sections from 12-week-old and 1-year-old male mdx and mdx5cv mice stained with an antibody to collagen III. Scale bar = 25 μm. D: Quantification of the percentage of area of the diaphragm section positive for collagen III immunostaining. Values are means ± SDs from 5 mice per group. No significant differences were found.
      Figure thumbnail gr5
      Figure 5Quantification of inflammation and myofiber membrane damage in wild-type, mdx, and mdx5cv mice. A: Representative micrographs of diaphragm (Dia) and quadriceps (Qua) muscle sections stained with an antibody against the immune cell marker CD45. Scale bar = 25 μm. B: Quantification of the percentage of area of the muscle section positive for CD45 immunostaining. Values are means ± SDs from 5 mice per group. C and D: Quantification of the amount of Evans Blue dye incorporation in the diaphragm (C) and quadriceps (D) muscles of mdx and mdx5cv mice at 12 weeks and 1 year of age. No significant differences were found between mdx and mdx5cv mice. Data represent mean ± SD (n = 4).

      Microdystrophin Expression in the Diaphragm Improves Resistance to Exercise-Induced Fatigue in mdx5cv Mice

      Microdystrophin expression in mdx mice has been shown to significantly improve muscle strength and rescue histopathology.
      • Rodino-Klapac L.R.
      • Janssen P.M.
      • Montgomery C.L.
      • Coley B.D.
      • Chicoine L.G.
      • Clark K.R.
      • Mendell J.R.
      A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy.
      • Yoshimura M.
      • Sakamoto M.
      • Ikemoto M.
      • Mochizuki Y.
      • Yuasa K.
      • Miyagoe-Suzuki Y.
      • Takeda S.
      AAV vector-mediated microdystrophin expression in a relatively small percentage of mdx myofibers improved the mdx phenotype.
      • Liu M.
      • Yue Y.
      • Harper S.Q.
      • Grange R.W.
      • Chamberlain J.S.
      • Duan D.
      Adeno-associated virus-mediated microdystrophin expression protects young mdx muscle from contraction-induced injury.
      • Rodino-Klapac L.R.
      • Montgomery C.L.
      • Bremer W.G.
      • Shontz K.M.
      • Malik V.
      • Davis N.
      • Sprinkle S.
      • Campbell K.J.
      • Sahenk Z.
      • Clark K.R.
      • Walker C.M.
      • Mendell J.R.
      • Chicoine L.G.
      Persistent expression of FLAG-tagged micro dystrophin in nonhuman primates following intramuscular and vascular delivery.
      • Harper S.Q.
      • Hauser M.A.
      • DelloRusso C.
      • Duan D.
      • Crawford R.W.
      • Phelps S.F.
      • Harper H.A.
      • Robinson A.S.
      • Engelhardt J.F.
      • Brooks S.V.
      • Chamberlain J.S.
      Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy.
      We sought to determine whether microdystrophin expression in mdx5cv mice could lead to measurable improvements in motor function. One-day-old mdx5cv pups were treated with AAV8.MCK.microdystrophin following a protocol shown to result in body-wide transduction.
      • Wang Z.
      • Zhu T.
      • Qiao C.
      • Zhou L.
      • Wang B.
      • Zhang J.
      • Chen C.
      • Li J.
      • Xiao X.
      Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart.
      Because sex cannot be established in neonates, female and male wild-type, control mdx5cv, and AAV8.MCK.microdystrophin mdx5cv mice were evaluated for exercise-induced fatigue and gait at 12 weeks of age and then sacrificed for analysis of microdystrophin expression in limb and diaphragm muscles.
      No effect of AAV8.MCK.microdystrophin treatment was detected on any gait parameter measured (stride length, paw separation, hind paw width), even after taking sex differences into account (data not shown). For exercise-induced fatigue, no significant sex differences were found, and data from both sexes were pooled. mdx5cv mice treated with AAV8.MCK.microdystrophin showed a significant improvement in exercise-induced fatigue compared with control mdx5cv mice for all parameters measured (Figure 6, A and B). As expected, values were below wild-type levels because microdystrophin does not restore neuronal nitric oxide synthase (nNOS) at the membrane of myofibers,
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      • Lai Y.
      • Thomas G.D.
      • Yue Y.
      • Yang H.T.
      • Li D.
      • Long C.
      • Judge L.
      • Bostick B.
      • Chamberlain J.S.
      • Terjung R.L.
      • Duan D.
      Dystrophins carrying spectrin-like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy.
      and this protein plays an important role in muscle fatigue.
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      Figure thumbnail gr6
      Figure 6Microdystrophin expression in the diaphragm of mdx5cv mice improves resistance to exercise-induced fatigue. A and B: Effects of microdystrophin expression in the diaphragm of mdx5cv mice on exercise-induced fatigue at 12 weeks of age. AAV8.MCK.microdystrophin mdx5cv mice show significant improvements for all activity parameters measured compared with mdx5cv mice but do not reach wild-type values. Significant differences among groups shown: *P < 0.05, **P < 0.001, and ***P < 0.0005. An analysis of variance was used for comparisons. C: Western blot analysis of microdystrophin protein expression in the diaphragm of wild-type and AAV8.MCK.microdystrophin mdx5cv mice. Actin is shown as a loading control.
      Histologic and Western blot analyses of muscles from AAV8.MCK.microdystrophin-treated mdx5cv mice showed that we achieved high transduction of the diaphragm in all injected mice (Figure 6C) and that microdystrophin was correctly localized to the myofiber membrane (see Supplemental Figure S6A at http://ajp.amjpathol.org). Furthermore, the percentage of fibers with centrally located nuclei (Table 1), the amount of connective tissue, and the incorporation of Evans Blue dye (see Supplemental Figure S6A at http://ajp.amjpathol.org) were visibly reduced. Interestingly, no microdystrophin expression was detected in limb muscles, possibly explaining the lack of improved gait in treated mice. Triceps, gastrocnemius, quadriceps, and tibialis anterior muscles showed large areas of Evans Blue dye-positive fibers (see Supplemental Figure S6B at http://ajp.amjpathol.org; data not shown), and most myofibers had centrally located nuclei, similar to control mdx5cv mice (data not shown).
      Table 1Comparison of Micro-Dystrophin Expression, Myofiber Regeneration, and Changes in Postexercise Activity in Individual AAV8.MCK.microdystrophin mdx5cv Mice
      Mouse ID%μDYS protein%DYS+fibers%FCNAmbulation (%)Rearing (%)Distance (%)Rest time (s)
      169512.6−85−89−841275
      2111714.7−93−91−942604
      320402.2−82−83−731189
      427308.8−66−77−64694
      530303.1−78−79−801471
      mdx5cv controls51 ± 5−91 ± 2−97 ± 1−90 ± 92029 ± 707
      Ambulation, rearing events, and distance travelled are expressed as percentage of change in activity after exercise. Rest time shown is postexercise value after subtraction of rest time value before exercise. Reference values for control mdx5cv mice represent mean ± SD.
      %μDYS, percentage of micro-dystrophin protein expression relative to dystrophin in wild-type diaphragm as determined by Western blot analysis (Figure 6C), and values were normalized to actin as a loading control for each lane; % DYS+fibers, percentage of micro-dystrophin positive myofibers per tissue sections; %FCN, percentage of fibers with central nuclei.
      Therefore, expression of microdystrophin in the diaphragm significantly improves resistance to exercise-induced fatigue in mdx5cv mice independent of nNOS restoration. Because microdystrophin expression was not detected in limb muscles, these results suggest that the diaphragm contributes significantly to the high sensitivity of mdx5cv mice to exercise-induced fatigue. They also indicate that this assay is robust and sensitive enough to detect treatment effects in small numbers of experimental animals.

      Discussion

      In this study, we report that the mdx5cv mouse, a commercially available model of DMD (The Jackson Laboratory stock no. 002379), has more severe functional deficits than the more widely used mdx mouse with respect to motor coordination, resistance to exercise-induced fatigue, and gait abnormalities. Furthermore, our study showed that the greater susceptibility of mdx5cv mice to exercise-induced fatigue primarily arises from a more severe force-generation deficit and enhanced susceptibility to contraction-induced fatigue of their diaphragm muscle compared with mdx mice. This conclusion is based on our ex vivo force measurements as well as on the in vivo finding that expression of microdystrophin in the diaphragm of mdx5cv mice significantly improves their resistance to exercise-induced fatigue. Notably, the beneficial effects of microdystrophin on fatigability in mdx5cv mice are probably independent of improved histopathology or nNOS signaling because transgenic expression of microdystrophin in mdx mice normalizes histologic parameters but does not protect from exercise-induced fatigue.
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      Overall, our results are consistent with the existence of a genetic modifier of the dystrophic phenotype in mdx5cv mice. mdx5cv mice were generated by chemical mutagenesis and backcrossed for 11 generations onto the C57BL/6 background (The Jackson Laboratory, personal communication), whereas mdx mice arose from a spontaneous mutation on the C57BL/10 strain. The C57BL/10 and C57BL/6 strains share a common ancestor and are generally considered interchangeable. However, genetic differences have been reported
      • McClive P.J.
      • Huang D.
      • Morahan G.
      C57BL/6 and C57BL/10 inbred mouse strains differ at multiple loci on chromosome 4.
      • Slingsby J.H.
      • Hogarth M.B.
      • Simpson E.
      • Walport M.J.
      • Morley B.J.
      New microsatellite polymorphisms identified between C57BL/6 C57BL/10, and C57BL/KsJ inbred mouse strains.
      as well as some behavioral differences.
      • Deacon R.M.
      • Thomas C.L.
      • Rawlins J.N.
      • Morley B.J.
      A comparison of the behavior of C57BL/6 and C57BL/10 mice.
      • Lewis S.R.
      • Ahmed S.
      • Dym C.
      • Khaimova E.
      • Kest B.
      • Bodnar R.J.
      Inbred mouse strain survey of sucrose intake.
      The effects of this genetic modifier appear silent in the absence of muscular dystrophy because we and others
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      • Deacon R.M.
      • Thomas C.L.
      • Rawlins J.N.
      • Morley B.J.
      A comparison of the behavior of C57BL/6 and C57BL/10 mice.
      • Rafael J.A.
      • Nitta Y.
      • Peters J.
      • Davies K.E.
      Testing of SHIRPA, a mouse phenotypic assessment protocol, on Dmd(mdx) and Dmd(mdx3cv) dystrophin-deficient mice.
      • Barton E.R.
      Impact of sarcoglycan complex on mechanical signal transduction in murine skeletal muscle.
      found no significant differences in motor performance or force measurements between wild-type C57BL/6 and C57BL/10 mice. Furthermore, it appears distinct from recently identified modifiers of the dystrophic phenotype that affect all skeletal muscles and typically result in increased fibrosis.
      • Fukada S.-i.
      • Morikawa D.
      • Yamamoto Y.
      • Yoshida T.
      • Sumie N.
      • Yamaguchi M.
      • Ito T.
      • Miyagoe-Suzuki Y.
      • Takeda Si
      • Tsujikawa K.
      • Yamamoto H.
      Genetic background affects properties of satellite cells and mdx phenotypes.
      • Heydemann A.
      • Ceco E.
      • Lim J.E.
      • Hadhazy M.
      • Ryder P.
      • Moran J.L.
      • Beier D.R.
      • Palmer A.A.
      • McNally E.M.
      Latent TGF-beta-binding protein 4 modifies muscular dystrophy in mice.
      • Chandrasekharan K.
      • Yoon J.H.
      • Xu Y.
      • deVries S.
      • Camboni M.
      • Janssen P.M.
      • Varki A.
      • Martin P.T.
      A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy.
      • Sacco A.
      • Mourkioti F.
      • Tran R.
      • Choi J.
      • Llewellyn M.
      • Kraft P.
      • Shkreli M.
      • Delp S.
      • Pomerantz J.H.
      • Artandi S.E.
      • Blau H.M.
      Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice.
      • Pegoraro E.
      • Hoffman E.P.
      • Piva L.
      • Gavassini B.F.
      • Cagnin S.
      • Ermani M.
      • Bello L.
      • Soraru G.
      • Pacchioni B.
      • Bonifati M.D.
      • Lanfranchi G.
      • Angelini C.
      • Kesari A.
      • Lee I.
      • Gordish-Dressman H.
      • Devaney J.M.
      • McDonald C.M.
      SPP1 genotype is a determinant of disease severity in Duchenne muscular dystrophy.
      Uniquely, the phenotype of mdx5cv mice appears linked to a mechanism that specifically affects the contractile properties of the diaphragm and can be corrected by microdystrophin independent of nNOS restoration. Interestingly, among skeletal muscles, the diaphragm had the largest number of differentially expressed genes between mdx5cv and mdx mice.
      • Haslett J.N.
      • Kang P.B.
      • Han M.
      • Kho A.T.
      • Sanoudou D.
      • Volinski J.M.
      • Beggs A.H.
      • Kohane I.S.
      • Kunkel L.M.
      The influence of muscle type and dystrophin deficiency on murine expression profiles.
      Further studies are needed to elucidate the mechanisms underlying a more severe diaphragm involvement in mdx5cv mice because they may be relevant to variability in the DMD phenotype in humans.
      The primary goal of this study was to determine whether mdx5cv mice have more severe motor function deficits than mdx mice, thus providing a mouse model of DMD better suited to study the effect of treatments on motor function and endurance. Currently, preclinical studies involving mdx mice primarily rely on indirect measures (histology and force measurements on a limited subset of muscles) to predict treatment effect on overall motor function and fatigability, which may not be accurate as shown for microdystrophin.
      • Kobayashi Y.M.
      • Rader E.P.
      • Crawford R.W.
      • Iyengar N.K.
      • Thedens D.R.
      • Faulkner J.A.
      • Parikh S.V.
      • Weiss R.M.
      • Chamberlain J.S.
      • Moore S.A.
      • Campbell K.P.
      Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
      Our results indicate that mdx5cv mice should be considered as an alternative to mdx mice for these studies for several reasons. First, because of the severity of their functional deficits, the performance of mdx5cv mice was strikingly inferior to that of wild-type mice, providing a large dynamic range both in terms of motor performance and age, over which treatment effects can be evaluated. This is not the case for the mdx mouse, whereby a motor performance deficit is only reliably detectable at older ages or is not detected at all as shown for the Rotarod assay. Second, mdx5cv mice show quantifiable and significant deficits at younger ages (typically 6 to 8 weeks) compared with mdx mice. Therefore, treatment effects can be assayed much earlier in mdx5cv mice and can be followed over longer periods of time. In addition, compounding effects of aging on phenotype can be avoided, especially for some tests such as gait analysis. Third, mdx5cv mice tend to show lower interindividual variability in motor activity tests compared with mdx mice. This is a significant advantage when planning pilot studies with small numbers of animals.
      Therefore, our results show that mdx5cv and mdx mice are not interchangeable models of DMD. Their differences hint at novel disease mechanisms that may influence disease outcome. Importantly, our findings indicate that the mdx5cv mouse model can significantly extend the utility of the mouse in DMD preclinical studies that want to include in vivo muscle function as an outcome measure without the compounding effects of exercise-induced muscle damage or old age.

      Acknowledgments

      We thank Drs. Jerry Mendell, Kevin Flanigan, and Carlos Miranda for their critical review of this manuscript. We also thank Dr. Chalonda Handy for help with the Rotarod Assay and Dr. Haiyan Fu for technical assistance with the Photobeam activity system.

      Supplementary data

      • Supplemental Figure S1

        Gait analysis tracings and measurements. A: Representative gait tracing from wild-type C57BL/6, mdx, and mdx5cv mice. Red: front paw; blue: hind paw. B: Hind paw base width (BW) measurement. C: Hind to front paw separation (S) measurement. D: Stride length (SL) measurement.

      • Supplemental Figure S3

        C57BL/10 and C57BL/6 wild-type mice have similar exercise-induced fatigue. Changes in ambulation, rearing events, distance traveled, and rest time after exercise. Values are normalized to pre-exercise levels. No significant differences were found between C57BL/10 and C57BL/6 wild-type mice. Data are mean ± SD. Values for 8 and 12 weeks are for male mice, whereas female mice were analyzed at the 1-year time point.

      • Supplemental Figure S4

        C57BL/10 and C57BL/6 wild-type mice have similar gait patterns. Measurements of stride length, hind paw to forepaw separation, and hind paw base width in C57BL/10 and C57BL/6 wild-type mice at 8 weeks, 12 weeks, and 1 year of age. Data represent mean ± SE.

      • Supplemental Figure S5

        Limb grip strength and EDL force measurements in wild-type, mdx, and mdx5cv mice. A: Raw grip strength measurements for hind limbs and forelimbs of 42-week-old mice. No significant difference was found between mdx and mdx5cv mice or between C57BL/10 and C57BL/6 wild-type mice. Data are mean ± SD (n = 5 to 6 mice). Significant differences from wild-type mice: *P < 0.05, ***P < 0.005. B: Tetanic force at optimal length of EDL muscles. The mdx and mdx5cv mice have a significant lower force development compared with strain-matched wild-type mice (P < 0.05, repeated-measures analysis of variance) but do not differ from each other. Force values were normalized to cross-sectional area and are underestimated by 30% to 40% because of light tissue blotting before weighing (see Materials and Methods). C: EDL muscle response to the repetitive eccentric contraction protocol. The mdx and mdx5cv mice show more severe loss of force during 10 repeats of lengthening contractions compared with wild-type controls (*P < 0.05). However, no significant difference was found between the mdx and mdx5cv mice.

      • Supplemental Figure S6

        Microdystrophin expression in the diaphragm of mdx5cv mice improves histopathology. A, top panel: Immunostaining of diaphragm tissue sections from 12-week-old wild-type (BL6), mdx5cv, and AAV8.MCK.microdystrophin mdx5cv (mdx5cv μDYS) mice with an antibody to dystrophin and microdystrophin (green). Fibers with a compromised membrane are stained with Evans Blue dye (red). Microdystrophin-expressing fibers have an intact membrane as indicated by the lack of Evans Blue dye staining (red). Nuclei are labeled blue. A, bottom panel: Diaphragm sections stained for hematoxylin/eosin show decreased connective tissue, inflammation, and necrotic foci in AAV8.MCK.microdystrophin-treated mdx5cv mice. B: Representative microdystrophin immunostaining (green) of a quadriceps muscle section showing lack of limb transduction in AAV8.MCK.microdystrophin mdx5cv mice and presence of fibers with a damaged membrane (red). Nuclei are labeled blue. Scale bars = 200 μm.

      References

        • Dubowitz V.
        Muscle Disorders in Childhood.
        W. B. Saunders, Philadelphia1995
        • Hoffman E.P.
        • Brown Jr, R.H.
        • Kunkel L.M.
        Dystrophin: the protein product of the Duchenne muscular dystrophy locus.
        Cell. 1987; 51: 919-928
        • Monaco A.P.
        • Neve R.L.
        • Colletti-Feener C.
        • Bertelson C.J.
        • Kurnit D.M.
        • Kunkel L.M.
        Isolation of candidate cDNAs for portions of the Duchenne muscular dystrophy gene.
        Nature. 1986; 323: 646-650
      1. Carpenter S. Karpati G. Disease of Skeletal Muscle. Oxford University Press Inc, New York, NY2001: 373-524
        • Matsumura K.
        • Ohlendieck K.
        • Ionasescu V.V.
        • Tome F.M.
        • Nonaka I.
        • Burghes A.H.
        • Mora M.
        • Kaplan J.C.
        • Fardeau M.
        • Campbell K.P.
        The role of the dystrophin-glycoprotein complex in the molecular pathogenesis of muscular dystrophies.
        Neuromuscul Disord. 1993; 3: 533-535
        • Engel A.G.
        • Ozawa E.
        Dystrophinopathies.
        in: Engel A.G. Franzini-Armstrong C. Myology. 2nd ed. McGraw-Hill, New York, NY2004: 961-1025 (AG, C)
        • Zhou L.
        • Lu H.
        Targeting fibrosis in Duchenne muscular dystrophy.
        J Neuropathol Exp Neurol. 2010; 69: 771-776
        • Mitrpant C.
        • Fletcher S.
        • Wilton S.D.
        Personalised genetic intervention for Duchenne muscular dystrophy: antisense oligomers and exon skipping.
        Curr Mol Pharmacol. 2009; 2: 110-121
        • Nelson S.F.
        • Crosbie R.H.
        • Miceli M.C.
        • Spencer M.J.
        Emerging genetic therapies to treat Duchenne muscular dystrophy.
        Current Opin Neurol. 2009; 22: 532-538
        • Muir L.A.
        • Chamberlain J.S.
        Emerging strategies for cell and gene therapy of the muscular dystrophies.
        Expert Rev Mol Med. 2009; 11: e18
        • Peault B.
        • Rudnicki M.
        • Torrente Y.
        • Cossu G.
        • Tremblay J.P.
        • Partridge T.
        • Gussoni E.
        • Kunkel L.M.
        • Huard J.
        Stem and progenitor cells in skeletal muscle development, maintenance, and therapy.
        Mol Ther. 2007; 15: 867-877
        • Cossu G.
        • Sampaolesi M.
        New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials.
        Trends Mol Med. 2007; 13: 520-526
        • Bulfield G.
        • Siller W.G.
        • Wight P.A.
        • Moore K.J.
        X chromosome-linked muscular dystrophy (mdx) in the mouse.
        Proc Natl Acad Sci U S A. 1984; 81: 1189-1192
        • McGeachie J.K.
        • Grounds M.D.
        • Partridge T.A.
        • Morgan J.E.
        Age-related changes in replication of myogenic cells in mdx mice: quantitative autoradiographic studies.
        J Neurol Sci. 1993; 119: 169-179
        • Pastoret C.
        • Sebille A.
        mdx mice show progressive weakness and muscle deterioration with age.
        J Neurol Sci. 1995; 129: 97-105
        • Lefaucheur J.P.
        • Pastoret C.
        • Sebille A.
        Phenotype of dystrophinopathy in old mdx mice.
        Anat Rec. 1995; 242: 70-76
        • Stedman H.H.
        • Sweeney H.L.
        • Shrager J.B.
        • Maguire H.C.
        • Panettieri R.A.
        • Petrof B.
        • Narusawa M.
        • Leferovich J.M.
        • Sladky J.T.
        • Kelly A.M.
        The mdx mouse diaphragm reproduces the degenerative changes of Duchenne muscular dystrophy.
        Nature. 1991; 352: 536-539
        • Chamberlain J.S.
        • Metzger J.
        • Reyes M.
        • Townsend D.
        • Faulkner J.A.
        Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma.
        FASEB J. 2007; 21 (2195-2104)
        • Carnwath J.W.
        • Shotton D.M.
        Muscular dystrophy in the mdx mouse: histopathology of the soleus and extensor digitorum longus muscles.
        J Neurol Sci. 1987; 80: 39-54
        • Tanabe Y.
        • Esaki K.
        • Nomura T.
        Skeletal muscle pathology in X chromosome-linked muscular dystrophy (mdx) mouse.
        Acta Neuropathol (Berl). 1986; 69: 91-95
        • Torres L.F.
        • Duchen L.W.
        The mutant mdx: inherited myopathy in the mouse.
        Brain. 1987; 110: 269-299
        • Dangain J.
        • Vrbova G.
        Muscle development in mdx mutant mice.
        Muscle Nerve. 1984; 7: 700-704
        • Muntoni F.
        • Mateddu A.
        • Marchei F.
        • Clerk A.
        • Serra G.
        Muscular weakness in the mdx mouse.
        J Neurol Sci. 1993; 120: 71-77
        • Grounds M.D.
        • Radley H.G.
        • Lynch G.S.
        • 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
        • Spurney C.F.
        • Gordish-Dressman H.
        • Guerron A.D.
        • Sali A.
        • Pandey G.S.
        • Rawat R.
        • Van Der Meulen J.H.
        • Cha H.J.
        • Pistilli E.E.
        • Partridge T.A.
        • Hoffman E.P.
        • Nagaraju K.
        Preclinical drug trials in the mdx mouse: assessment of reliable and sensitive outcome measures.
        Muscle Nerve. 2009; 39: 591-602
        • Chapman V.M.
        • Miller D.R.
        • Armstrong D.
        • Caskey C.T.
        Recovery of Induced Mutations for X Chromosome-Linked Muscular Dystrophy in Mice.
        Proc Natl Acad Sci U S A. 1989; 86: 1292-1296
        • Araki E.
        • Nakamura K.
        • Nakao K.
        • Kameya S.
        • Kobayashi O.
        • Nonaka I.
        • Kobayashi T.
        • Katsuki M.
        Targeted disruption of exon 52 in the mouse dystrophin gene induced muscle degeneration similar to that observed in Duchenne muscular dystrophy.
        Biochem Biophys Res Commun. 1997; 238: 492-497
        • Fukada S.-i.
        • Morikawa D.
        • Yamamoto Y.
        • Yoshida T.
        • Sumie N.
        • Yamaguchi M.
        • Ito T.
        • Miyagoe-Suzuki Y.
        • Takeda Si
        • Tsujikawa K.
        • Yamamoto H.
        Genetic background affects properties of satellite cells and mdx phenotypes.
        Am J Pathol. 2010; 176: 2414-2424
        • Im W.B.
        • Phelps S.F.
        • Copen E.H.
        • Adams E.G.
        • Slightom J.L.
        • Chamberlain J.S.
        Differential expression of dystrophin isoforms in strains of mdx mice with different mutations.
        Hum Mol Genet. 1996; 5: 1149-1153
        • Danko I.
        • Chapman V.
        • Wolff J.A.
        The frequency of revertants in mdx mouse genetic models for Duchenne muscular dystrophy.
        Pediatr Res. 1992; 32: 128-131
        • Haslett J.N.
        • Kang P.B.
        • Han M.
        • Kho A.T.
        • Sanoudou D.
        • Volinski J.M.
        • Beggs A.H.
        • Kohane I.S.
        • Kunkel L.M.
        The influence of muscle type and dystrophin deficiency on murine expression profiles.
        Mamm Genome. 2005; 16: 739-748
        • Payne E.T.
        • Yasuda N.
        • Bourgeois J.M.
        • Devries M.C.
        • Rodriguez M.C.
        • Yousuf J.
        • Tarnopolsky M.A.
        Nutritional therapy improves function and complements corticosteroid intervention in mdx mice.
        Muscle Nerve. 2006; 33: 66-77
        • Kobayashi Y.M.
        • Rader E.P.
        • Crawford R.W.
        • Iyengar N.K.
        • Thedens D.R.
        • Faulkner J.A.
        • Parikh S.V.
        • Weiss R.M.
        • Chamberlain J.S.
        • Moore S.A.
        • Campbell K.P.
        Sarcolemma-localized nNOS is required to maintain activity after mild exercise.
        Nature. 2008; 456: 511-515
        • Carter R.J.
        • Lione L.A.
        • Humby T.
        • Mangiarini L.
        • Mahal A.
        • Bates G.P.
        • Dunnett S.B.
        • Morton A.J.
        Characterization of progressive motor deficits in mice transgenic for the human Huntington's disease mutation.
        J Neurosci. 1999; 19: 3248-3257
        • Hamer P.W.
        • McGeachie J.M.
        • Davies M.J.
        • Grounds M.D.
        Evans Blue Dye as an in vivo marker of myofibre damage: optimising parameters for detecting initial myofibre membrane permeability.
        J Anat. 2002; 200: 69-79
        • Heydemann A.
        • Ceco E.
        • Lim J.E.
        • Hadhazy M.
        • Ryder P.
        • Moran J.L.
        • Beier D.R.
        • Palmer A.A.
        • McNally E.M.
        Latent TGF-beta-binding protein 4 modifies muscular dystrophy in mice.
        J Clin Invest. 2009; 119: 3703-3712
        • Zhou L.
        • Rafael-Fortney J.A.
        • Huang P.
        • Zhao X.S.
        • Cheng G.
        • Zhou X.
        • Kaminski H.J.
        • Liu L.
        • Ransohoff R.M.
        Haploinsufficiency of utrophin gene worsens skeletal muscle inflammation and fibrosis in mdx mice.
        J Neurol Sci. 2008; 264: 106-111
        • Huang P.
        • Zhao X.S.
        • Fields M.
        • Ransohoff R.M.
        • Zhou L.
        Imatinib attenuates skeletal muscle dystrophy in mdx mice.
        FASEB J. 2009; 23: 2539-2548
        • Reddy G.K.
        • Enwemeka C.S.
        A simplified method for the analysis of hydroxyproline in biological tissues.
        Clin Biochem. 1996; 29: 225-229
        • Acharyya S.
        • Villalta S.A.
        • Bakkar N.
        • Bupha-Intr T.
        • Janssen P.M.
        • Carathers M.
        • Li Z.W.
        • Beg A.A.
        • Ghosh S.
        • Sahenk Z.
        • Weinstein M.
        • Gardner K.L.
        • Rafael-Fortney J.A.
        • Karin M.
        • Tidball J.G.
        • Baldwin A.S.
        • Guttridge D.C.
        Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy.
        J Clin Invest. 2007; 117: 889-901
        • Chandrasekharan K.
        • Yoon J.H.
        • Xu Y.
        • deVries S.
        • Camboni M.
        • Janssen P.M.
        • Varki A.
        • Martin P.T.
        A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy.
        Sci Transl Med. 2010; 2 (42ra54)
        • Peterson J.M.
        • Kline W.
        • Canan B.D.
        • Ricca D.J.
        • Kaspar B.
        • Delfin D.A.
        • Dirienzo K.
        • Clemens P.R.
        • Robbins P.D.
        • Baldwin A.S.
        • Flood P.
        • Kaumaya P.
        • Frietas M.
        • Kornegay J.N.
        • Mendell J.R.
        • Rafael-Fortney J.A.
        • Guttridge D.C.
        • Janssen P.M.
        Peptide-based inhibition of NF-kappaB rescues diaphragm muscle contractile dysfunction in a murine model of Duchenne muscular dystrophy.
        Mol Med. 2011; 17: 508-515
        • Rucker M.
        • Fraites Jr, T.J.
        • Porvasnik S.L.
        • Lewis M.A.
        • Zolotukhin I.
        • Cloutier D.A.
        • Byrne B.J.
        Rescue of enzyme deficiency in embryonic diaphragm in a mouse model of metabolic myopathy: pompe disease.
        Development. 2004; 131: 3007-3019
        • Diaz P.T.
        • Brownstein E.
        • Clanton T.L.
        Effects of N-acetylcysteine on in vitro diaphragm function are temperature dependent.
        J Appl Physiol. 1994; 77: 2434-2439
        • Prezant D.J.
        • Richner B.
        • Valentine D.E.
        • Aldrich T.K.
        • Fishman C.L.
        • Nagashima H.
        • Chaudhry I.
        • Cahill J.
        Temperature dependence of rat diaphragm muscle contractility and fatigue.
        J Appl Physiol. 1990; 69: 1740-1745
        • Rodino-Klapac L.R.
        • Janssen P.M.
        • Montgomery C.L.
        • Coley B.D.
        • Chicoine L.G.
        • Clark K.R.
        • Mendell J.R.
        A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy.
        J Transl Med. 2007; 5: 45
        • Martin P.T.
        • Xu R.
        • Rodino-Klapac L.R.
        • Oglesbay E.
        • Camboni M.
        • Montgomery C.L.
        • Shontz K.
        • Chicoine L.
        • Clark K.R.
        • Sahenk Z.
        • Mendell J.R.
        • Janssen P.M.
        Overexpression of Galgt2 in skeletal muscle prevents injury resulting from eccentric contractions in both mdx and wild type mice.
        Am J Physiol Cell Physiol. 2009; 296: C476-C488
        • Rabinowitz J.E.
        • Rolling F.
        • Li C.
        • Conrath H.
        • Xiao W.
        • Xiao X.
        • Samulski R.J.
        Cross-packaging of a single adeno-associated virus (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity.
        J Virol. 2002; 76: 791-801
        • Hauser M.A.
        • Amalfitano A.
        • Kumar-Singh R.
        • Hauschka S.D.
        • Chamberlain J.S.
        Improved adenoviral vectors for gene therapy of Duchenne muscular dystrophy.
        Neuromuscul Disord. 1997; 7: 277-283
        • Cohn R.D.
        • Liang H.Y.
        • Shetty R.
        • Abraham T.
        • Wagner K.R.
        Myostatin does not regulate cardiac hypertrophy or fibrosis.
        Neuromuscul Disord. 2007; 17: 290-296
        • Hayes A.
        • Williams D.A.
        Contractile function and low-intensity exercise effects of old dystrophic (mdx) mice.
        Am J Physiol. 1998; 274: C1138-C1144
        • Hooper A.C.
        Bone length and muscle weight in mice subjected to genetic selection for the relative length of the tibia and radius.
        Life Sci. 1978; 22: 283-286
        • Golumbek P.T.
        • Keeling R.M.
        • Connolly A.M.
        Strength and corticosteroid responsiveness of mdx mice is unchanged by RAG2 gene knockout.
        Neuromuscul Disord. 2007; 17: 376-384
        • Wang B.
        • Li J.
        • Fu F.H.
        • Xiao X.
        Systemic human minidystrophin gene transfer improves functions and life span of dystrophin and dystrophin/utrophin-deficient mice.
        J Orthop Res. 2009; 27: 421-426
        • Zeman R.J.
        • Peng H.
        • Danon M.J.
        • Etlinger J.D.
        Clenbuterol reduces degeneration of exercised or aged dystrophic (mdx) muscle.
        Muscle Nerve. 2000; 23: 521-528
        • Wooddell C.I.
        • Zhang G.
        • Griffin J.B.
        • Hegge J.O.
        • Huss T.
        • Wolff J.A.
        Use of Evans blue dye to compare limb muscles in exercised young and old mdx mice.
        Muscle Nerve. 2010; 41: 487-499
        • Straub V.
        • Rafael J.A.
        • Chamberlain J.S.
        • Campbell K.P.
        Animal models for muscular dystrophy show different patterns of sarcolemmal disruption.
        J Cell Biol. 1997; 139: 375-385
        • Yoshimura M.
        • Sakamoto M.
        • Ikemoto M.
        • Mochizuki Y.
        • Yuasa K.
        • Miyagoe-Suzuki Y.
        • Takeda S.
        AAV vector-mediated microdystrophin expression in a relatively small percentage of mdx myofibers improved the mdx phenotype.
        Mol Ther. 2004; 10: 821-828
        • Liu M.
        • Yue Y.
        • Harper S.Q.
        • Grange R.W.
        • Chamberlain J.S.
        • Duan D.
        Adeno-associated virus-mediated microdystrophin expression protects young mdx muscle from contraction-induced injury.
        Mol Ther. 2005; 11: 245-256
        • Rodino-Klapac L.R.
        • Montgomery C.L.
        • Bremer W.G.
        • Shontz K.M.
        • Malik V.
        • Davis N.
        • Sprinkle S.
        • Campbell K.J.
        • Sahenk Z.
        • Clark K.R.
        • Walker C.M.
        • Mendell J.R.
        • Chicoine L.G.
        Persistent expression of FLAG-tagged micro dystrophin in nonhuman primates following intramuscular and vascular delivery.
        Mol Ther. 2010; 18: 109-117
        • Harper S.Q.
        • Hauser M.A.
        • DelloRusso C.
        • Duan D.
        • Crawford R.W.
        • Phelps S.F.
        • Harper H.A.
        • Robinson A.S.
        • Engelhardt J.F.
        • Brooks S.V.
        • Chamberlain J.S.
        Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy.
        Nat Med. 2002; 8: 253-261
        • Wang Z.
        • Zhu T.
        • Qiao C.
        • Zhou L.
        • Wang B.
        • Zhang J.
        • Chen C.
        • Li J.
        • Xiao X.
        Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart.
        Nature Biotechnol. 2005; 23: 321-328
        • Lai Y.
        • Thomas G.D.
        • Yue Y.
        • Yang H.T.
        • Li D.
        • Long C.
        • Judge L.
        • Bostick B.
        • Chamberlain J.S.
        • Terjung R.L.
        • Duan D.
        Dystrophins carrying spectrin-like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy.
        J Clin Invest. 2009; 119: 624-635
        • McClive P.J.
        • Huang D.
        • Morahan G.
        C57BL/6 and C57BL/10 inbred mouse strains differ at multiple loci on chromosome 4.
        Immunogenetics. 1994; 39: 286-288
        • Slingsby J.H.
        • Hogarth M.B.
        • Simpson E.
        • Walport M.J.
        • Morley B.J.
        New microsatellite polymorphisms identified between C57BL/6.
        Immunogenetics. 1996; 43: 72-75
        • Deacon R.M.
        • Thomas C.L.
        • Rawlins J.N.
        • Morley B.J.
        A comparison of the behavior of C57BL/6 and C57BL/10 mice.
        Behav Brain Res. 2007; 179: 239-247
        • Lewis S.R.
        • Ahmed S.
        • Dym C.
        • Khaimova E.
        • Kest B.
        • Bodnar R.J.
        Inbred mouse strain survey of sucrose intake.
        Physiol Behav. 2005; 85: 546-556
        • Rafael J.A.
        • Nitta Y.
        • Peters J.
        • Davies K.E.
        Testing of SHIRPA, a mouse phenotypic assessment protocol, on Dmd(mdx) and Dmd(mdx3cv) dystrophin-deficient mice.
        Mamm Genome. 2000; 11: 725-728
        • Barton E.R.
        Impact of sarcoglycan complex on mechanical signal transduction in murine skeletal muscle.
        Am J Physiol Cell Physiol. 2006; 290: C411-C419
        • Sacco A.
        • Mourkioti F.
        • Tran R.
        • Choi J.
        • Llewellyn M.
        • Kraft P.
        • Shkreli M.
        • Delp S.
        • Pomerantz J.H.
        • Artandi S.E.
        • Blau H.M.
        Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice.
        Cell. 2010; 143: 1059-1071
        • Pegoraro E.
        • Hoffman E.P.
        • Piva L.
        • Gavassini B.F.
        • Cagnin S.
        • Ermani M.
        • Bello L.
        • Soraru G.
        • Pacchioni B.
        • Bonifati M.D.
        • Lanfranchi G.
        • Angelini C.
        • Kesari A.
        • Lee I.
        • Gordish-Dressman H.
        • Devaney J.M.
        • McDonald C.M.
        SPP1 genotype is a determinant of disease severity in Duchenne muscular dystrophy.
        Neurology. 2011; 76: 219-226