VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects

Christopher R Heier, Jesse M Damsker, Qing Yu, Blythe C Dillingham, Tony Huynh, Jack H Van der Meulen, Arpana Sali, Brittany K Miller, Aditi Phadke, Luana Scheffer, James Quinn, Kathleen Tatem, Sarah Jordan, Sherry Dadgar, Olga C Rodriguez, Chris Albanese, Michael Calhoun, Heather Gordish-Dressman, Jyoti K Jaiswal, Edward M Connor, John M McCall, Eric P Hoffman, Erica K M Reeves, Kanneboyina Nagaraju, Christopher R Heier, Jesse M Damsker, Qing Yu, Blythe C Dillingham, Tony Huynh, Jack H Van der Meulen, Arpana Sali, Brittany K Miller, Aditi Phadke, Luana Scheffer, James Quinn, Kathleen Tatem, Sarah Jordan, Sherry Dadgar, Olga C Rodriguez, Chris Albanese, Michael Calhoun, Heather Gordish-Dressman, Jyoti K Jaiswal, Edward M Connor, John M McCall, Eric P Hoffman, Erica K M Reeves, Kanneboyina Nagaraju

Abstract

Absence of dystrophin makes skeletal muscle more susceptible to injury, resulting in breaches of the plasma membrane and chronic inflammation in Duchenne muscular dystrophy (DMD). Current management by glucocorticoids has unclear molecular benefits and harsh side effects. It is uncertain whether therapies that avoid hormonal stunting of growth and development, and/or immunosuppression, would be more or less beneficial. Here, we discover an oral drug with mechanisms that provide efficacy through anti-inflammatory signaling and membrane-stabilizing pathways, independent of hormonal or immunosuppressive effects. We find VBP15 protects and promotes efficient repair of skeletal muscle cells upon laser injury, in opposition to prednisolone. Potent inhibition of NF-κB is mediated through protein interactions of the glucocorticoid receptor, however VBP15 shows significantly reduced hormonal receptor transcriptional activity. The translation of these drug mechanisms into DMD model mice improves muscle strength, live-imaging and pathology through both preventive and post-onset intervention regimens. These data demonstrate successful improvement of dystrophy independent of hormonal, growth, or immunosuppressive effects, indicating VBP15 merits clinical investigation for DMD and would benefit other chronic inflammatory diseases.

Keywords: anti-inflammatory; dystrophy; mdx; membrane injury; muscle.

© 2013 The Authors. Published by John Wiley and Sons, Ltd on behalf of EMBO.

Figures

Figure 1. VBP15 structure differentiates it from…
Figure 1. VBP15 structure differentiates it from glucocorticoids and improves GR specificity
  1. A–D. The chemical structure of VBP15 is provided (A). VBP15 was selected for clinical development by having the optimal profile from a library of Δ-9,11 compounds, whose general structure is provided in (B). Compound diversity for this library was generated by medicinal chemistry probing of the R1–R3 groups. These compounds are structurally related to glucocorticoids (C), but contain an essential Δ-9,11 double bond modification to the steroid C-ring, at the location depicted by the red box. This modification produces novel properties and clear differences in sub-activity profiles of these compounds. Prednisolone (D) is the active form of prednisone, a current glucocorticoid standard of care for DMD.

  2. E–H. Receptor specificity was determined through competitive binding assays. Here, radiolabeled high-affinity ligands were incubated with extracted steroid receptors and increasing concentrations of unlabeled competitor (high-affinity control, VBP15, VBP1 or VBP3). Best-fit curves are provided. VBP15 showed increased GR specificity through increased binding to the (E) GR and decreased binding to the (F) MR in comparison to other Δ-9,11 compounds (VBP1 and VBP3). VBP15 also showed low (H) androgen receptor binding and no detectable binding to the (G) oestrogen receptor. (Triam, triamcinolone; Spiro, spironolactone; R1881, methyltrienolone; Estradiol, 17β-estradiol).

Figure 2. VBP15 inhibits inflammatory signaling and…
Figure 2. VBP15 inhibits inflammatory signaling and promotes membrane stability
  1. A. In an NF-κB reporter assay, increasing concentrations of prednisolone or VBP15 were applied to TNFα-induced C2C12 myoblasts stably expressing a luciferase reporter under the control of an NF-κB driven promoter. Significant reporter inhibition was observed at all concentrations over 10 nM.

  2. B. Inhibition was also observed for endogenous NF-κB activated inflammatory transcripts. Cox2, Irf1 and Nos2 expression was significantly reduced in TNFα-induced H2K myotubes, as determined by real time qPCR. Results are mean ± SEM from representative experiments performed in triplicate (ANOVA, *p < 0.05, **p < 0.005, ***p < 0.0005).

  3. C,D. Laser injury and dye exclusion assays to determine effects of VBP15 and prednisolone on cell membrane integrity. (C) Images of C2C12 myoblasts exposed to the indicated drug 15 min prior to laser wounding, with fluorescent visualization of FM1-43 dye entry into cells (white arrowheads mark sites of injury, scale bars = 5 µm). VBP15 shows protection against laser-induced injury. (D) Quantitation of FM1-43 influx over time (laser injury at time 6 s). VBP15 shows reduced impact of initial injury and enhanced repair, whereas prednisolone shows greater impact from injury with elevated dye uptake. Representative data from one of four experiments are presented as mean ± SEM. (ANOVA, n ≥ 16 per treatment, *p < 0.05; Pred, prednisolone).

Figure 3. GR is required for anti-inflammatory…
Figure 3. GR is required for anti-inflammatory activities of VBP15 and prednisolone, but VBP15 shows loss of GRE-mediated sub-activities associated with side effects
  1. A. Application of a steroidal receptor antagonist (RU-486) ablated the NF-κB inhibitory activity of both prednisolone and VBP15, indicating that both drugs share steroidal anti-inflammatory pathways.

  2. B,C. Assessment of GRE-mediated transcriptional (hormonal) activities differentiates prednisolone and VBP15. (B) Sgk1 gene expression is controlled by a positive GRE and showed reduced activation by VBP15 in comparison to glucocorticoids in AtT-20 pituitary cells, as measured by qRT-PCR. α-Tocopherol was included as a negative control compound that lacks any GRE activity. (C) ACTH expression is controlled by a negative GRE, and is considered a component of adrenal suppression (negative side effect of pharmacological glucocorticoids). VBP15 showed reduced effects on this side effect pathway via ELISA of treated AtT-20 cell media.

  3. D. Western blot of GRnull fibroblasts and the control GR positive L929 line they were derived from, illustrating the absence of detectable GR protein in the GRnull cells. GAPDH was included as a loading control.

  4. E,F.GRnull and GR positive fibroblasts were treated with drug, then induced with TNFα and transcript levels assayed by real time qPCR. (E) Sgk1 levels illustrate an absence of induction in GRnull cells, confirming Sgk1 dependence on the GR and the absence of GR function in this cell line. (F) Inhibition of the endogenous NF-κB activated inflammatory transcripts Irf1, Tnfα and Il1a was observed in GR positive cells but not in GRnull cells, indicating the GR is essential for this inhibition.

  5. G,H. Spleens were harvested from GRdim/dim and wild type control mice. Splenocyte suspensions were treated with drug, then induced with TNFα and transcripts assayed by qPCR. (G) Nfkbia levels illustrate an absence of GRE induction by the mutant GR, as well as a lack of induction of NF-κB inhibiting gene products, in GRdim/dim splenocytes. (H) Inhibition of Irf1, Tnfα and Il6 inflammatory transcripts was observed in both wild type and GRdim/dim mutant splenocytes. This indicates GRdim isoforms, which maintain protein-protein interactions but lose receptor-DNA interactions, still maintain inhibition of endogenous NF-κB activated inflammatory transcripts. (Pred, prednisolone; Dex, dexamethasone; α-Toc, vitamin E; ANOVA, *p < 0.05, **p < 0.005, ***p < 0.0005).

Figure 4. VBP15 improves dystrophic phenotypes of…
Figure 4. VBP15 improves dystrophic phenotypes of mdx mice in two pre-clinical trials (pre-symptomatic and post-onset treatment regimens)
  1. A–F. Prophylactic treatment of mdx mice beginning at 2 weeks of age showed dose-dependent improvement of clinical and histological endpoints. Mouse limb strength increased upon VBP15 treatment as measured by grip strength of 6 week old mice for both (A) forelimb and (B) hindlimb (n ≥ 12 mice/group). (C) Maximal force exerted by mouse forelimbs increased with VBP15 treatment but decreased with prednisolone treatment, due to prednisolone effects on mouse size (presented later). (D) Specific force of isolated EDL muscle increased with VBP15 treatment (n = 10 mice/group). (E) Live-animal imaging of cathepsin protease activity (ProSense680) shows reduced inflammation and necrosis of the hindlimbs in VBP15-treated mdx mice (E images; E′ quantitation of fluorescence; n ≥ 6 mice/group). (F) Histology of diaphragm muscle shows a decrease in inflammatory foci from VBP15 treatment at 15 and 30 mg/kg (F representative images, F′ quantitation; n = 6 mice/group).

  2. G–I. A second pre-clinical trial was performed in exercised adult mdx mice to assay post-onset efficacy. (G) Live-animal imaging of inflammation (ProSense680) showed a significant decrease with VBP15 treatment (G representative images, (G′) quantitation; n ≥ 6 mice/group). (H) Specific force of isolated EDL muscle was measured ex vivo at trial conclusion with 15 mg/kg VBP15 showing an increase consistent with the neonate trial (n ≥ 7 mice/group). (I) Histology of adult diaphragm showed a significant reduction in inflammatory foci upon 45 mg/kg VBP15 treatment (n = 6 mice/group). Values are mean ± SEM. For treatments, the mean percentage of increase or decrease of mdx vehicle values towards WT is provided. (Pred, prednisolone; FL, forelimb; HL, hindlimb; data exceeding 2 SD's was removed from specific force values as an outlier but included in all statistical analyses; one-tailed t-test of single dose versus vehicle mdxp < 0.05; ANOVA of dose-dependence groups versus vehicle mdx *p < 0.05, **p < 0.005, ***p < 0.0005).

Figure 5. VBP15 does not show immunosuppressive…
Figure 5. VBP15 does not show immunosuppressive activities shown by prednisolone
  1. Prednisolone significantly reduced the number of viable splenocytes per gram of spleen tissue, whereas VBP15 did not at any dose.

  2. The percentage of B lymphocytes, as measured by FACS analysis of B220 positive cells, was reduced in spleens from prednisolone treated mdx mice, while VBP15 showed no decrease in B cells.

  3. Spleen CD4+ T cell numbers were significantly decreased in prednisolone treated mdx spleens, but not by VBP15 treatment.

  4. Activation of mdx splenocyte T cells by concavalin A was impaired by prednisolone, but not impaired by VBP15 treatment. Values are mean ± SEM. (Pred, prednisolone; (A) n ≥ 12, (B–D) n = 3–5; *p ≤ 0.05, **p < 0.005).

Figure 6. VBP15 lacks the side effects…
Figure 6. VBP15 lacks the side effects of current glucocorticoid regimens in vivo
  1. A. Prednisolone treatment stunted the growth of developing mice in comparison to both vehicle and VBP15 groups. Representative photographs (A) and quantitation of body length (A′) are provided.

  2. B. Bone lengths were reduced upon prednisolone treatment. X-rays (B) of mouse tibias illustrate size differences (scale bars = 2 mm). Quantitation shows a significant decrease in tibia length (B′).

  3. C. MicroCT imaging analysis of femur revealed a significant decrease in trabecular thickness (C′) for prednisolone treated mice.

  4. D–F. Increases in cardiac fibrosis and heart mass were detected in prednisolone treated mice, suggestive of cardiac damage as a side effect lacking for VBP15. Sirius red staining of cardiac muscle shows increased fibrosis in prednisolone-treated mice, but not VBP15 mice. Representative images (D) and digital quantitation of fibrosis (E) are provided. To the right of the image panel is a higher magnification image from the area outlined in box. (F) Heart mass ratios were increased by prednisolone but not by VBP15.

  5. G,H. In adult mdx mice as well, increases in cardiac fibrosis (G) and heart mass (G) were observed with prednisolone treatment but not VBP15 treatment. In adult mdx vehicle mice, an expected disease- and age-related increase in fibrosis over WT is seen. Values are mean ± SEM. (n ≥ 12 per group for (A,B,E,F); n ≥ 5 for (C,G,H); *p < 0.05, **p < 0.005, ***p < 0.0005).

Figure 7. Working model of VBP15 and…
Figure 7. Working model of VBP15 and prednisone drug mechanism sub-activity profiles
Steroidal compounds such as glucocorticoids (prednisone) and Δ-9,11 compounds (VBP15) are multi-potent drugs. Through dissecting the sub-activities of these compounds, we find that: (1) VBP15 reduces inflammation but does not show the immunosuppressive impairment of lymphocyte viability and function observed for prednisone. (2) Within an environment of plasma membrane disruption, VBP15 helps to promote resistance to and repair of injuries, while prednisone can exacerbate membrane injury. (3) Inside cells, both compounds bind to and activate the GR to potently inhibit inflammatory NF-κB signaling through protein–protein interactions. (4) Though they both bind to the GR, prednisone causes strong induction of hormonal GRE controlled promoter elements, while VBP15 eliminates or greatly reduces these effects.

References

    1. Aoki Y, Nakamura A, Yokota T, Saito T, Okazawa H, Nagata T, Takeda S. In-frame dystrophin following exon 51-skipping improves muscle pathology and function in the exon 52-deficient mdx mouse. Mol Ther. 2010;18:1995–2005.
    1. Avioli LV. Glucocorticoid effects on statural growth. Br J Rheumatol. 1993;32:27–30.
    1. Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, McNeil PL, Campbell KP. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature. 2003;423:168–172.
    1. Baudy AR, Saxena N, Gordish H, Hoffman EP, Nagaraju K. A robust in vitro screening assay to identify NF-kappaB inhibitors for inflammatory muscle diseases. Int Immunopharmacol. 2009;9:1209–1214.
    1. Baudy AR, Sali A, Jordan S, Kesari A, Johnston HK, Hoffman EP, Nagaraju K. Non-invasive optical imaging of muscle pathology in mdx mice using cathepsin caged near-infrared imaging. Mol Imaging Biol. 2011;13:462–470.
    1. Baudy AR, Reeves EK, Damsker JM, Heier C, Garvin LM, Dillingham BC, McCall J, Rayavarapu S, Wang Z, Vandermeulen JH, et al. Delta-9,11 modification of glucocorticoids dissociates nuclear factor-kappaB inhibitory efficacy from glucocorticoid response element-associated side effects. J Pharmacol Exp Ther. 2012;343:225–232.
    1. Bauer R, Straub V, Blain A, Bushby K, MacGowan GA. Contrasting effects of steroids and angiotensin-converting-enzyme inhibitors in a mouse model of dystrophin-deficient cardiomyopathy. Eur J Heart Fail. 2009;11:463–471.
    1. Bazan NG, Marcheselli VL, Cole-Edwards K. Brain response to injury and neurodegeneration: endogenous neuroprotective signaling. Ann N Y Acad Sci. 2005;1053:137–147.
    1. Bertorini TE, Bhattacharya SK, Palmieri GM, Chesney CM, Pifer D, Baker B. Muscle calcium and magnesium content in Duchenne muscular dystrophy. Neurology. 1982;32:1088–1092.
    1. Bircan Z, Soran M, Yildirim I, Dogan M, Sahin A, Bilici A, Danaci M. The effect of alternate-day low dose prednisolone on bone age in children with steroid dependent nephrotic syndrome. Int Urol Nephrol. 1997;29:357–361.
    1. Brooks SV, Faulkner JA. Contractile properties of skeletal muscles from young, adult and aged mice. J Physiol. 1988;404:71–82.
    1. Burkin DJ, Wuebbles RD. A molecular bandage for diseased muscle. Sci Transl Med. 2012;4:139fs119.
    1. Buttgereit F, Brand MD, Burmester GR. Equivalent doses and relative drug potencies for non-genomic glucocorticoid effects: a novel glucocorticoid hierarchy. Biochem Pharmacol. 1999;58:363–368.
    1. Cai D, Frantz JD, Tawa NE, Jr, Melendez PA, Oh BC, Lidov HG, Hasselgren PO, Frontera WR, Lee J, Glass DJ, et al. IKKbeta/NF-kappaB activation causes severe muscle wasting in mice. Cell. 2004;119:285–298.
    1. Chase TH, Cox GA, Burzenski L, Foreman O, Shultz LD. Dysferlin deficiency and the development of cardiomyopathy in a mouse model of limb-girdle muscular dystrophy 2B. Am J Pathol. 2009;175:2299–2308.
    1. Chen YW, Nagaraju K, Bakay M, McIntyre O, Rawat R, Shi R, Hoffman EP. Early onset of inflammation and later involvement of TGFbeta in Duchenne muscular dystrophy. Neurology. 2005;65:826–834.
    1. Dahlman-Wright K, Wright A, Gustafsson JA, Carlstedt-Duke J. Interaction of the glucocorticoid receptor DNA-binding domain with DNA as a dimer is mediated by a short segment of five amino acids. J Biol Chem. 1991;266:3107–3112.
    1. Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter AC, Metzinger L, Watt DJ, Dickson JG, Tinsley JM, Davies KE. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell. 1997;90:717–727.
    1. Delfin DA, Xu Y, Peterson JM, Guttridge DC, Rafael-Fortney JA, Janssen PM. Improvement of cardiac contractile function by peptide-based inhibition of NF-kappaB in the utrophin/dystrophin-deficient murine model of muscular dystrophy. J Transl Med. 2011;9:68.
    1. Diamond MI, Miner JN, Yoshinaga SK, Yamamoto KR. Transcription factor interactions: selectors of positive or negative regulation from a single DNA element. Science. 1990;249:1266–1272.
    1. Disatnik MH, Dhawan J, Yu Y, Beal MF, Whirl MM, Franco AA, Rando TA. Evidence of oxidative stress in mdx mouse muscle: studies of the pre-necrotic state. J Neurol Sci. 1998;161:77–84.
    1. Drouin J, Sun YL, Chamberland M, Gauthier Y, De Lean A, Nemer M, Schmidt TJ. Novel glucocorticoid receptor complex with DNA element of the hormone-repressed POMC gene. EMBO J. 1993;12:145–156.
    1. Dumonceaux J, Marie S, Beley C, Trollet C, Vignaud A, Ferry A, Butler-Browne G, Garcia L. Combination of myostatin pathway interference and dystrophin rescue enhances tetanic and specific force in dystrophic mdx mice. Mol Ther. 2010;18:881–887.
    1. Gajic O, Lee J, Doerr CH, Berrios JC, Myers JL, Hubmayr RD. Ventilator-induced cell wounding and repair in the intact lung. Am J Respir Crit Care Med. 2003;167:1057–1063.
    1. Gehrig SM, van der Poel C, Sayer TA, Schertzer JD, Henstridge DC, Church JE, Lamon S, Russell AP, Davies KE, Febbraio MA, et al. Hsp72 preserves muscle function and slows progression of severe muscular dystrophy. Nature. 2012;484:394–398.
    1. Goyenvalle A, Babbs A, Powell D, Kole R, Fletcher S, Wilton SD, Davies KE. Prevention of dystrophic pathology in severely affected dystrophin/utrophin-deficient mice by morpholino-oligomer-mediated exon-skipping. Mol Ther. 2010;18:198–205.
    1. Grady RM, Merlie JP, Sanes JR. Subtle neuromuscular defects in utrophin-deficient mice. J Cell Biol. 1997a;136:871–882.
    1. Grady RM, Teng H, Nichol MC, Cunningham JC, Wilkinson RS, Sanes JR. Skeletal and cardiac myopathies in mice lacking utrophin and dystrophin: a model for Duchenne muscular dystrophy. Cell. 1997b;90:729–738.
    1. Grady RM, Grange RW, Lau KS, Maimone MM, Nichol MC, Stull JT, Sanes JR. Role for alpha-dystrobrevin in the pathogenesis of dystrophin-dependent muscular dystrophies. Nat Cell Biol. 1999;1:215–220.
    1. Granchelli JA, Avosso DL, Hudecki MS, Pollina C. Cromolyn increases strength in exercised mdx mice. Res Commun Mol Pathol Pharmacol. 1996;91:287–296.
    1. Griggs RC, Moxley RT, III, Mendell JR, Fenichel GM, Brooke MH, Pestronk A, Miller JP, Cwik VA, Pandya S, Robison J, et al. Duchenne dystrophy: randomized, controlled trial of prednisone (18 months) and azathioprine (12 months) Neurology. 1993;43:520–527.
    1. Grounds MD, Shavlakadze T. Growing muscle has different sarcolemmal properties from adult muscle: a proposal with scientific and clinical implications: reasons to reassess skeletal muscle molecular dynamics, cellular responses and suitability of experimental models of muscle disorders. Bioessays. 2011;33:458–468.
    1. Grounds MD, Torrisi J. Anti-TNFalpha (Remicade) therapy protects dystrophic skeletal muscle from necrosis. FASEB J. 2004;18:676–682.
    1. Guo C, Willem M, Werner A, Raivich G, Emerson M, Neyses L, Mayer U. Absence of alpha 7 integrin in dystrophin-deficient mice causes a myopathy similar to Duchenne muscular dystrophy. Hum Mol Genet. 2006;15:989–998.
    1. Heine VM, Rowitch DH. Hedgehog signaling has a protective effect in glucocorticoid-induced mouse neonatal brain injury through an 11betaHSD2-dependent mechanism. J Clin Invest. 2009;119:267–277.
    1. Hoffman EP, Bronson A, Levin AA, Takeda S, Yokota T, Baudy AR, Connor EM. Restoring dystrophin expression in duchenne muscular dystrophy muscle progress in exon skipping and stop codon read through. Am J Pathol. 2011;179:12–22.
    1. Housley PR, Forsthoefel AM. Isolation and characterization of a mouse L cell variant deficient in glucocorticoid receptors. Biochem Biophys Res Commun. 1989;164:480–487.
    1. Howard AC, McNeil AK, McNeil PL. Promotion of plasma membrane repair by vitamin E. Nat Commun. 2011;2:597.
    1. Itani OA, Liu KZ, Cornish KL, Campbell JR, Thomas CP. Glucocorticoids stimulate human sgk1 gene expression by activation of a GRE in its 5′-flanking region. Am J Physiol Endocrinol Metab. 2002;283:E971–E979.
    1. Jaiswal JK, Marlow G, Summerill G, Mahjneh I, Mueller S, Hill M, Miyake K, Haase H, Anderson LV, Richard I, et al. Patients with a non-dysferlin Miyoshi myopathy have a novel membrane repair defect. Traffic. 2007;8:77–88.
    1. Johnsen SD. Prednisone therapy in Becker's muscular dystrophy. J Child Neurol. 2001;16:870–871.
    1. Kar NC, Pearson CM. Muscular dystrophy and activation of proteinases. Muscle Nerve. 1978;1:308–313.
    1. Kavanagh RJ, Kam PC. Lazaroids: efficacy and mechanism of action of the 21-aminosteroids in neuroprotection. Br J Anaesth. 2001;86:110–119.
    1. Kissel JT, Lynn DJ, Rammohan KW, Klein JP, Griggs RC, Moxley RT, III, Cwik VA, Brooke MH, Mendell JR. Mononuclear cell analysis of muscle biopsies in prednisone- and azathioprine-treated Duchenne muscular dystrophy. Neurology. 1993;43:532–536.
    1. Knuesel I, Riban V, Zuellig RA, Schaub MC, Grady RM, Sanes JR, Fritschy JM. Increased vulnerability to kainate-induced seizures in utrophin-knockout mice. Eur J Neurosci. 2002;15:1474–1484.
    1. Landis SC, Amara SG, Asadullah K, Austin CP, Blumenstein R, Bradley EW, Crystal RG, Darnell RB, Ferrante RJ, Fillit H, et al. A call for transparent reporting to optimize the predictive value of preclinical research. Nature. 2012;490:187–191.
    1. Lipworth BJ. Therapeutic implications of non-genomic glucocorticoid activity. Lancet. 2000;356:87–89.
    1. Manolagas SC, Weinstein RS. New developments in the pathogenesis and treatment of steroid-induced osteoporosis. J Bone Miner Res. 1999;14:1061–1066.
    1. Marques MJ, Ventura Machado R, Minatel E, Santo Neto H. Disodium cromoglycate protects dystrophin-deficient muscle fibers from leakiness. Muscle Nerve. 2008;37:61–67.
    1. Megeney LA, Kablar B, Perry RL, Ying C, May L, Rudnicki MA. Severe cardiomyopathy in mice lacking dystrophin and MyoD. Proc Natl Acad Sci USA. 1999;96:220–225.
    1. Meijsing SH, Pufall MA, So AY, Bates DL, Chen L, Yamamoto KR. DNA binding site sequence directs glucocorticoid receptor structure and activity. Science. 2009;324:407–410.
    1. Mourkioti F, Kratsios P, Luedde T, Song YH, Delafontaine P, Adami R, Parente V, Bottinelli R, Pasparakis M, Rosenthal N. Targeted ablation of IKK2 improves skeletal muscle strength, maintains mass, and promotes regeneration. J Clin Invest. 2006;116:2945–2954.
    1. Nagaraju K, Willmann R. Developing standard procedures for murine and canine efficacy studies of DMD therapeutics: report of two expert workshops on “Pre-clinical testing for Duchenne dystrophy”: Washington DC, October 27th–28th 2007 and Zurich, June 30th–July 1st 2008. Neuromuscul Disord. 2009;19:502–506.
    1. Peterson JM, Kline W, Canan BD, Ricca DJ, Kaspar B, Delfin DA, DiRienzo K, Clemens PR, Robbins PD, Baldwin AS, et al. 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.
    1. Pinsky L, Digeorge AM. Cleft palate in the mouse: a teratogenic index of glucocorticoid potency. Science. 1965;147:402–403.
    1. Porter JD, Khanna S, Kaminski HJ, Rao JS, Merriam AP, Richmonds CR, Leahy P, Li J, Guo W, Andrade FH. A chronic inflammatory response dominates the skeletal muscle molecular signature in dystrophin-deficient mdx mice. Hum Mol Genet. 2002;11:263–272.
    1. Porter JD, Merriam AP, Leahy P, Gong B, Khanna S. Dissection of temporal gene expression signatures of affected and spared muscle groups in dystrophin-deficient (mdx) mice. Hum Mol Genet. 2003;12:1813–1821.
    1. Rando TA, Disatnik MH, Yu Y, Franco A. Muscle cells from mdx mice have an increased susceptibility to oxidative stress. Neuromuscul Disord. 1998;8:14–21.
    1. Raymackers JM, Debaix H, Colson-Van Schoor M, De Backer F, Tajeddine N, Schwaller B, Gailly P, Gillis JM. Consequence of parvalbumin deficiency in the mdx mouse: histological, biochemical and mechanical phenotype of a new double mutant. Neuromuscul Disord. 2003;13:376–387.
    1. Reeves EK, Hoffman EP, Nagaraju K, Damsker JM, McCall JM. VBP15: preclinical characterization of a novel anti-inflammatory delta 9,11 steroid. Bioorg Med Chem. 2013;21:2241–2249.
    1. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, Bock R, Gass P, Schmid W, Herrlich P, Angel P, et al. DNA binding of the glucocorticoid receptor is not essential for survival. Cell. 1998;93:531–541.
    1. Rhen T, Grissom S, Afshari C, Cidlowski JA. Dexamethasone blocks the rapid biological effects of 17beta-estradiol in the rat uterus without antagonizing its global genomic actions. FASEB J. 2003;17:1849–1870.
    1. Saija A, Tomaino A, Pellegrino ML, Giuffrida N, Trombetta D, Castelli F. In vitro evaluation of the antioxidant activity and biomembrane interaction of the lazaroid U-74389G. Life Sci. 2001;68:1351–1366.
    1. Sali A, Guerron AD, Gordish-Dressman H, Spurney CF, Iantorno M, Hoffman EP, Nagaraju K. Glucocorticoid-treated mice are an inappropriate positive control for long-term preclinical studies in the mdx mouse. PLoS ONE. 2012;7:e34204.
    1. Sharma N, Medikayala S, Defour A, Rayavarapu S, Brown KJ, Hathout Y, Jaiswal JK. Use of quantitative membrane proteomics identifies a novel role of mitochondria in healing injured muscles. J Biol Chem. 2012;287:30455–30467.
    1. Shivaji S, Jagannadham MV. Steroid-induced perturbations of membranes and its relevance to sperm acrosome reaction. Biochim Biophys Acta. 1992;1108:99–109.
    1. Spurney CF, Gordish-Dressman H, Guerron AD, Sali A, Pandey GS, Rawat R, Van Der Meulen JH, Cha HJ, Pistilli EE, Partridge TA, et al. Preclinical drug trials in the mdx mouse: assessment of reliable and sensitive outcome measures. Muscle Nerve. 2009;39:591–602.
    1. Spurney CF, Guerron AD, Yu Q, Sali A, van der Meulen JH, Hoffman EP, Nagaraju K. Membrane sealant Poloxamer P188 protects against isoproterenol induced cardiomyopathy in dystrophin deficient mice. BMC Cardiovasc Disord. 2011;11:20.
    1. Takeda A, Jimi T, Wakayama Y, Misugi N, Miyake S, Kumagai T. Demonstration of cathepsins B, H and L in xenografts of normal and Duchenne-muscular-dystrophy muscles transplanted into nude mice. Biochem J. 1992;288:643–648.
    1. Townsend D, Turner I, Yasuda S, Martindale J, Davis J, Shillingford M, Kornegay JN, Metzger JM. Chronic administration of membrane sealant prevents severe cardiac injury and ventricular dilatation in dystrophic dogs. J Clin Invest. 2010;120:1140–1150.
    1. Wakefield PM, Tinsley JM, Wood MJ, Gilbert R, Karpati G, Davies KE. Prevention of the dystrophic phenotype in dystrophin/utrophin-deficient muscle following adenovirus-mediated transfer of a utrophin minigene. Gene Ther. 2000;7:201–204.
    1. Weisleder N, Takizawa N, Lin P, Wang X, Cao C, Zhang Y, Tan T, Ferrante C, Zhu H, Chen PJ, et al. Recombinant MG53 protein modulates therapeutic cell membrane repair in treatment of muscular dystrophy. Sci Transl Med. 2012;4:139ra185.
    1. Willmann R, Possekel S, Dubach-Powell J, Meier T, Ruegg MA. Mammalian animal models for Duchenne muscular dystrophy. Neuromuscul Disord. 2009;19:241–249.
    1. Willmann R, De Luca A, Benatar M, Grounds M, Dubach J, Raymackers JM, Nagaraju K. Enhancing translation: guidelines for standard pre-clinical experiments in mdx mice. Neuromuscul Disord. 2012;22:43–449.
    1. Wissink S, van Heerde EC, Schmitz ML, Kalkhoven E, van der Burg B, Baeuerle PA, van der Saag PT. Distinct domains of the RelA NF-kappaB subunit are required for negative cross-talk and direct interaction with the glucocorticoid receptor. J Biol Chem. 1997;272:22278–22284.
    1. Wolthers OD, Pedersen S. Short term linear growth in asthmatic children during treatment with prednisolone. BMJ. 1990;301:145–148.
    1. Yasuda S, Townsend D, Michele DE, Favre EG, Day SM, Metzger JM. Dystrophic heart failure blocked by membrane sealant poloxamer. Nature. 2005;436:1025–1029.
    1. Yoshiuchi I, Shingu R, Nakajima H, Hamaguchi T, Horikawa Y, Yamasaki T, Oue T, Ono A, Miyagawa JI, Namba M, et al. Mutation/polymorphism scanning of glucose-6-phosphatase gene promoter in noninsulin-dependent diabetes mellitus patients. J Clin Endocrinol Metab. 1998;83:1016–1019.

Source: PubMed

3
구독하다