Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy

Christopher R Heier, Qing Yu, Alyson A Fiorillo, Christopher B Tully, Asya Tucker, Davi A Mazala, Kitipong Uaesoontrachoon, Sadish Srinivassane, Jesse M Damsker, Eric P Hoffman, Kanneboyina Nagaraju, Christopher F Spurney, Christopher R Heier, Qing Yu, Alyson A Fiorillo, Christopher B Tully, Asya Tucker, Davi A Mazala, Kitipong Uaesoontrachoon, Sadish Srinivassane, Jesse M Damsker, Eric P Hoffman, Kanneboyina Nagaraju, Christopher F Spurney

Abstract

Cardiomyopathy is a leading cause of death for Duchenne muscular dystrophy. Here, we find that the mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) can share common ligands but play distinct roles in dystrophic heart and skeletal muscle pathophysiology. Comparisons of their ligand structures indicate that the Δ9,11 modification of the first-in-class drug vamorolone enables it to avoid interaction with a conserved receptor residue (N770/N564), which would otherwise activate transcription factor properties of both receptors. Reporter assays show that vamorolone and eplerenone are MR antagonists, whereas prednisolone is an MR agonist. Macrophages, cardiomyocytes, and CRISPR knockout myoblasts show vamorolone is also a dissociative GR ligand that inhibits inflammation with improved safety over prednisone and GR-specific deflazacort. In mice, hyperaldosteronism activates MR-driven hypertension and kidney phenotypes. We find that genetic dystrophin loss provides a second hit for MR-mediated cardiomyopathy in Duchenne muscular dystrophy model mice, as aldosterone worsens fibrosis, mass and dysfunction phenotypes. Vamorolone successfully prevents MR-activated phenotypes, whereas prednisolone activates negative MR and GR effects. In conclusion, vamorolone targets dual nuclear receptors to treat inflammation and cardiomyopathy with improved safety.

Conflict of interest statement

ReveraGen BioPharma owns the method of use intellectual property relating to the use of Δ9,11 compounds to treat disease. EP Hoffman and K Nagaraju are co-founders of ReveraGen with shares in the company. JM Damsker is an employee of the company. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

© 2019 Heier et al.

Figures

Figure 1.. Vamorolone and MR antagonists lack…
Figure 1.. Vamorolone and MR antagonists lack 11-β hydroxyl groups linked to MR activation.
(A) Table of pharmacological and physiological MR ligands with their carbon 11 group identity provided. (B) Progesterone is a potent MR antagonist, whereas addition of an 11β-hydroxy (11β-Hydroxyprogesterone) results in an agonist compound. (C) The 11β-hydroxy group of hydroxyprogesterone interacts with MR residue N770 via hydrogen bonding. Dexamethasone also interacts with this conserved residue in the GR (N564) via hydrogen bonding. (D) Vamorolone is a Δ9,11 steroid where the 11β position features a carbon–carbon double bond, whereas prednisolone is an 11β-hydroxysteroid. (E) A stable MR reporter cell line was treated with drugs and quantified via chemiluminescence assay to determine their agonist properties. Prednisolone and aldosterone showed MR agonist activity. (F) Reporter cells were treated with drug in combination with a constant E80 dose of aldosterone to determine antagonist properties. Vamorolone acted as an MR antagonist, consistent with eplerenone and spironolactone. (Representative data from three experiments with each dose performed in triplicate; values are mean ± SEM).
Figure S1.. An active metabolite of deflazacort…
Figure S1.. An active metabolite of deflazacort shows no activity in MR reporter assays.
To determine if an active metabolite of deflazacort showed a lack of MR activity consistent with deflazacort, stable MR reporter cells were treated with 21-desacetyl deflazacort, prednisolone, or spironolactone. Used as controls here, prednisolone activated the MR as an agonist, and spironolactone acted as an antagonist to inhibit MR induction by aldosterone. Consistent with deflazacort, we found that the metabolite 21-desacetyl deflazacort avoided MR interaction. (A) Agonist activity was assayed by treating cells with each drug alone in increasing concentrations. (B) Antagonist activity was assayed by pretreating cells with each drug, then adding aldosterone (natural MR agonist) and assaying MR activity 6 h later. (Pred = Prednisolone, 21-Defl = 21-Desacetyl deflazacort, Spiro = Spironolactone).
Figure 2.. CRISPR knockout of the GR…
Figure 2.. CRISPR knockout of the GR shows that anti-inflammatory pathways are GR-dependent.
A frame-shifting deletion mutation was introduced via CRISPR in C2C12 myoblasts to knockout GR expression. (A) The DNA sequence of the GR N-terminus is provided. Base pairs deleted in knockout cells are shown as red, lower case letters. Positions of CRISPR guide RNAs are highlighted in yellow, and genotyping primers in blue. (B) PCR of C2C12 myoblast clones (labeled A through F) identified WT (∼500 bp band) and GR deletion clones (∼300 bp band). (C) Western blot confirmed GR knockout in clone from PCR lane F. (D) Drug-treated wild-type and GR knockout myoblasts were assayed for activation of a GR transactivation target transcript (Gilz). (E) Wild-type and GR knockout myotubes were treated with drug, induced with TNF, and assayed for expression of inflammatory genes regulated by NF-κB (Irf1 and Mcp1). (n = 3–4, *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001, ANOVA with post hoc versus vehicle [panel D] or + TNF [panel E] control; representative of three experiments; panels [D, E]: [−] = no drug or TNF, [+] = TNF plus vehicle, D = deflazacort, P = prednisolone, V = vamorolone, E = eplerenone).
Figure S2.. Vamorolone inhibits inflammatory signaling in…
Figure S2.. Vamorolone inhibits inflammatory signaling in macrophage cells.
RAW 264.7 macrophages were pretreated with 10 μM of GR and/or MR ligands and induced with LPS. Expression of NF-κB regulated inflammatory genes (Irf1 and Mcp1) was assayed by qRT-PCR. (n = 4, **P < 0.01, ***P < 0.005, ****P < 0.0001, ANOVA with post hoc versus [+] TNF control in gray; [−] = uninduced, [+] = TNF plus vehicle, D = deflazacort, P = prednisolone, V = vamorolone, E = eplerenone).
Figure 3.. Vamorolone inhibits inflammatory signaling in…
Figure 3.. Vamorolone inhibits inflammatory signaling in macrophage and heart cells.
(A) RAW 264.7 macrophages were pretreated with drug at 10 μM and inflammatory signaling was induced for 24 h using LPS. Expression of NF-κB–regulated inflammatory genes (Il1b and Il6) was assayed by qRT-PCR. (B) IL1B and IL6 protein levels were assayed in media from the same experiment via AlphaLISA assay. (C) Primary cardiomyocytes were pretreated with vehicle, vamorolone, or the GR-specific ligand deflazacort, and inflammatory signaling induced with TNF. NF-κB–regulated inflammatory gene expression (Il1b and Il6) was assayed by qRT-PCR. (D) IL6 protein levels were assayed by AlphaLISA. (E) HL-1cells were pretreated with 10 μM drug and induced with TNF for 24 h. Expression of Il6 was assayed by qRT-PCR. (n = 4, **P < 0.005, ****P < 0.0001, ANOVA with post hoc versus [+] TNF control in gray; [−] = no TNF control, [+] = TNF plus vehicle, D = deflazacort, P = prednisolone, V = vamorolone, E = eplerenone).
Figure 4.. Vamorolone is an effective MR…
Figure 4.. Vamorolone is an effective MR antagonist in vivo.
Mice were treated with daily oral drug (vehicle, vamorolone, eplerenone, or spironolactone) and implanted with osmotic pumps that secreted either aldosterone or vehicle for 6 wk. Kidney size was assayed as an epithelial MR target tissue, where aldosterone-specificity of the MR is maintained through expression of the glucocorticoid-inactivating enzyme HSD11B2. (A) Representative kidney images are provided. (B) Aldosterone induced an increase in kidney mass in both wild-type and mdx. This was prevented in mdx by all three MR antagonists. (n ≥ 8 per group; ***P < 0.0005, ANOVA with post hoc test versus mdx + aldosterone in red; ##P < 0.005, t test comparing WT groups; V = Vamorolone, E = Eplerenone, S = Spironolactone).
Figure 5.. Vamorolone improves MR-mediated hypertension and…
Figure 5.. Vamorolone improves MR-mediated hypertension and mdx cardiomyopathy phenotypes.
Aldosterone introduced via osmotic pumps caused an activation or progression of several phenotypes relevant to cardiomyopathy that were all improved or prevented by vamorolone, eplerenone, and spironolactone. (A) Blood pressure was measured by tail cuff, and increased by aldosterone in both wild-type and mdx mice. (B, C) Sirius Red staining of collagen deposition was used to assay heart fibrosis, where aldosterone increased heart fibrosis specifically in mdx mice in (B) representative images and upon (C) quantification. (D) qRT-PCR of mdx heart tissue was used to assay MR-activated gene expression changes relevant to fibrosis and cardiomyopathy (n = 6 per group). (E, F) Aldosterone increased mdx heart size in (E) representative images and (F) quantitative measurement of heart mass. (G–I) Heart function was assayed by echocardiography, with (G) fractional shortening (H) left ventricular wall thickness (LV wall thickness, in millimetre) and (I) left ventricular mass (LV mass) each showing aldosterone-mediated phenotypes that improved with antagonist treatment. (n ≥ 8; †P < 0.1, *P < 0.05, **P < 0.005, ***P < 0.0005, ANOVA with post hoc versus mdx + aldosterone in red; #P < 0.05, ##P < 0.005, t test comparing WT groups; V = Vamorolone, E = Eplerenone, S = Spironolactone).
Figure 6.. Prednisolone activates MR and GR…
Figure 6.. Prednisolone activates MR and GR side effect pathways in mdx and severe D2-mdx mice.
(A) Expression of the glucocorticoid-metabolizing gene Hsd11b2 was assayed by qRT-PCR in kidneys and hearts from wild-type and mdx mice. Lack of substantial expression in the heart indicated that the heart is sensitive to MR activation by prednisolone. (B) Standard mdx mice on a BL10 background were treated with vehicle, prednisolone (5 mg/kg), or vamorolone (45 mg/kg) for 4 months, beginning at 6 weeks of age. The heart was assayed for expression of MR-regulated genes via qRT-PCR and expressed as % vehicle levels (n ≥ 5). (C–G) Severe D2-mdx mice on an LTBP4 modifier background were treated for 8 weeks with daily oral vehicle, vamorolone (30 mg/kg), or prednisolone (5 mg/kg), beginning at 2 mo of age (n ≥ 9). (C, D) Kidney and heart mass at trial conclusion. Note, kidney expresses the glucocorticoid-inactivating enzyme (HSD11B2) that protects it from prednisolone, whereas the heart does not. Prednisolone increased D2-mdx heart size but not kidney size. (E, F) Echocardiography was performed near the trial endpoint. (E) Fractional shortening increased with both vamorolone and prednisolone. (F) Left ventricular volume (end diastolic) did not change with vamorolone but increased with prednisolone, consistent with premature dilatation. (G) Serum insulin levels increased with prednisolone (n ≥ 5), consistent with GR-mediated, metabolic side effects of glucocorticoids. (*P < 0.05, **P < 0.005, ***P < 0.0005; one panel B outlier removed after significant Grubbs test; (A) t test of mdx versus wild-type for each tissue, (B–G) ANOVA with post hoc versus vehicle in gray).
Figure 7.. Working model of dual-receptor drug…
Figure 7.. Working model of dual-receptor drug mechanisms in epithelial and non-epithelial tissues.
Left Panel: Epithelial MR target tissues (e.g., kidney, colon, skin) co-express the HSD11B2 enzyme to prevent over-activation of the MR by glucocorticoids. This protein metabolizes 11β-hydroxysteroids (cortisol and prednisolone) into their inactive ketone states (cortisone and prednisone) within that tissue. Aldosterone is not metabolized by HSD11B2, thus enabling aldosterone-specific signaling in vivo. We find that aldosterone challenge increases kidney size, whereas prednisolone treatment does not, consistent with this model. Right Panel: Non-epithelial MR target tissues express the MR but do not express the HSD11B2 enzyme (heart, brain, skin, fat, immune cells). These tissues may have increased risk of damage from MR activation by glucocorticoids. The heart is particularly important here, as (1) DMD patients naturally develop cardiomyopathy, and (2) a “second hit” provided by the DMD disease could worsen MR-mediated heart pathologies (such a “second hit” is seen in other aldosterone models). We find that both aldosterone and prednisolone increase mdx heart size and fibrosis, consistent with increased MR activation. Both Panels: Vamorolone is a potent MR antagonist that protects both epithelial and non-epithelial tissue types. (Black = physiological ligands, Blue = Vamorolone, Red = Prednisolone; Aldo = Aldosterone, Cort = Cortisol, HSD11B2 = 11β-hydroxysteroid dehydrogenase, Pred = Prednisolone, Vam = Vamorolone).

References

    1. Bauer R, Straub V, Blain A, Bushby K, MacGowan GA (2009) Contrasting effects of steroids and angiotensin-converting-enzyme inhibitors in a mouse model of dystrophin-deficient cardiomyopathy. Eur J Heart Fail 11: 463–471. 10.1093/eurjhf/hfp028
    1. Bello L, Gordish-Dressman H, Morgenroth LP, Henricson EK, Duong T, Hoffman EP, Cnaan A, McDonald CM (2015a) Prednisone/prednisolone and deflazacort regimens in the CINRG Duchenne Natural History Study. Neurology 85: 1048–1055. 10.1212/wnl.0000000000001950
    1. Bello L, Kesari A, Gordish-Dressman H, Cnaan A, Morgenroth LP, Punetha J, Duong T, Henricson EK, Pegoraro E, McDonald CM, et al. (2015b) Genetic modifiers of ambulation in the Cooperative International Neuromuscular Research Group Duchenne Natural History Study. Ann Neurol 77: 684–696. 10.1002/ana.24370
    1. Bienvenu LA, Morgan J, Rickard AJ, Tesch GH, Cranston GA, Fletcher EK, Delbridge LM, Young MJ (2012) Macrophage mineralocorticoid receptor signaling plays a key role in aldosterone-independent cardiac fibrosis. Endocrinology 153: 3416–3425. 10.1210/en.2011-2098
    1. Bledsoe RK, Montana VG, Stanley TB, Delves CJ, Apolito CJ, McKee DD, Consler TG, Parks DJ, Stewart EL, Willson TM, et al. (2002) Crystal structure of the glucocorticoid receptor ligand binding domain reveals a novel mode of receptor dimerization and coactivator recognition. Cell 110: 93–105. 10.1016/s0092-8674(02)00817-6
    1. Bonifati MD, Ruzza G, Bonometto P, Berardinelli A, Gorni K, Orcesi S, Lanzi G, Angelini C (2000) A multicenter, double-blind, randomized trial of deflazacort versus prednisone in Duchenne muscular dystrophy. Muscle Nerve 23: 1344–1347. 10.1002/1097-4598(200009)23:9<1344::aid-mus4>;2-6
    1. Brilla CG, Weber KT (1992) Mineralocorticoid excess, dietary sodium, and myocardial fibrosis. J Lab Clin Med 120: 893–901
    1. Bujak M, Frangogiannis NG (2009) The role of IL-1 in the pathogenesis of heart disease. Arch Immunol Ther Exp (Warsz) 57: 165–176. 10.1007/s00005-009-0024-y
    1. Chadwick JA, Bhattacharya S, Lowe J, Weisleder N, Rafael-Fortney JA (2017) Renin-angiotensin-aldosterone system inhibitors improve membrane stability and change gene-expression profiles in dystrophic skeletal muscles. Am J Physiol Cell Physiol 312: C155–C168. 10.1152/ajpcell.00269.2016
    1. Chadwick JA, Swager SA, Lowe J, Welc SS, Tidball JG, Gomez-Sanchez CE, Gomez-Sanchez EP, Rafael-Fortney JA (2016) Myeloid cells are capable of synthesizing aldosterone to exacerbate damage in muscular dystrophy. Hum Mol Genet 25: 5167–5177. 10.1093/hmg/ddw331
    1. Chen YW, Nagaraju K, Bakay M, McIntyre O, Rawat R, Shi R, Hoffman EP (2005) Early onset of inflammation and later involvement of TGFbeta in Duchenne muscular dystrophy. Neurology 65: 826–834. 10.1212/01.wnl.0000173836.09176.c4
    1. Claycomb WC, Lanson NA Jr., Stallworth BS, Egeland DB, Delcarpio JB, Bahinski A, Izzo NJ Jr (1998) HL-1 cells: A cardiac muscle cell line that contracts and retains phenotypic characteristics of the adult cardiomyocyte. Proc Natl Acad Sci U S A 95: 2979–2984. 10.1073/pnas.95.6.2979
    1. Cole TJ, Young MJ (2017) 30 years of the mineralocorticoid receptor: Mineralocorticoid receptor null mice: Informing cell-type-specific roles. J Endocrinol 234: T83–T92. 10.1530/joe-17-0155
    1. Coley WD, Bogdanik L, Vila MC, Yu Q, Van Der Meulen JH, Rayavarapu S, Novak JS, Nearing M, Quinn JL, Saunders A, et al. (2016) Effect of genetic background on the dystrophic phenotype in mdx mice. Hum Mol Genet 25: 130–145. 10.1093/hmg/ddv460
    1. Conklin LS, Damsker JM, Hoffman EP, Jusko WJ, Mavroudis PD, Schwartz BD, Mengle-Gaw LJ, Smith EC, Mah JK, Guglieri M, et al. (2018) Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first-in-class dissociative steroidal anti-inflammatory drug. Pharmacol Res 136: 140–150. 10.1016/j.phrs.2018.09.007
    1. Damsker JM, Conklin LS, Sadri S, Dillingham BC, Panchapakesan K, Heier CR, McCall JM, Sandler AD (2016) VBP15, a novel dissociative steroid compound, reduces NFkappaB-induced expression of inflammatory cytokines in vitro and symptoms of murine trinitrobenzene sulfonic acid-induced colitis. Inflamm Res 65: 737–743. 10.1007/s00011-016-0956-8
    1. Damsker JM, Dillingham BC, Rose MC, Balsley MA, Heier CR, Watson AM, Stemmy EJ, Jurjus RA, Huynh T, Tatem K, et al. (2013) VBP15, a glucocorticoid analogue, is effective at reducing allergic lung inflammation in mice. PLoS One 8: e63871 10.1371/journal.pone.0063871
    1. Danialou G, Comtois AS, Dudley R, Karpati G, Vincent G, Des Rosiers C, Petrof BJ (2001) Dystrophin-deficient cardiomyocytes are abnormally vulnerable to mechanical stress-induced contractile failure and injury. FASEB J 15: 1655–1657. 10.1096/fj.01-0030fje
    1. de Gasparo M, Joss U, Ramjoue HP, Whitebread SE, Haenni H, Schenkel L, Kraehenbuehl C, Biollaz M, Grob J, Schmidlin J, et al. (1987) Three new epoxy-spirolactone derivatives: Characterization in vivo and in vitro. J Pharmacol Exp Ther 240: 650–656
    1. de Gasparo M, Whitebread SE, Preiswerk G, Jeunemaitre X, Corvol P, Menard J (1989) Antialdosterones: Incidence and prevention of sexual side effects. J Steroid Biochem 32: 223–227. 10.1016/0022-4731(89)90169-6
    1. Di Zhang A, Nguyen Dinh Cat A, Soukaseum C, Escoubet B, Cherfa A, Messaoudi S, Delcayre C, Samuel JL, Jaisser F (2008) Cross-talk between mineralocorticoid and angiotensin II signaling for cardiac remodeling. Hypertension 52: 1060–1067. 10.1161/hypertensionaha.108.117531
    1. Diederich S, Eigendorff E, Burkhardt P, Quinkler M, Bumke-Vogt C, Rochel M, Seidelmann D, Esperling P, Oelkers W, Bahr V (2002) 11beta-hydroxysteroid dehydrogenase types 1 and 2: An important pharmacokinetic determinant for the activity of synthetic mineralo- and glucocorticoids. J Clin Endocrinol Metab 87: 5695–5701. 10.1210/jc.2002-020970
    1. Dillingham BC, Knoblach SM, Many GM, Harmon BT, Mullen AM, Heier CR, Bello L, McCall JM, Hoffman EP, Connor EM, et al. (2015) VBP15, a novel anti-inflammatory, is effective at reducing the severity of murine experimental autoimmune encephalomyelitis. Cell Mol Neurobiol 35: 377–387. 10.1007/s10571-014-0133-y
    1. Eagle M, Baudouin SV, Chandler C, Giddings DR, Bullock R, Bushby K (2002) Survival in Duchenne muscular dystrophy: Improvements in life expectancy since 1967 and the impact of home nocturnal ventilation. Neuromuscul Disord 12: 926–929. 10.1016/s0960-8966(02)00140-2
    1. Edwards CR, Stewart PM, Burt D, Brett L, McIntyre MA, Sutanto WS, de Kloet ER, Monder C (1988) Localisation of 11 beta-hydroxysteroid dehydrogenase--tissue specific protector of the mineralocorticoid receptor. Lancet 2: 986–989. 10.1016/s0140-6736(88)90742-8
    1. Ferraris JR, Pasqualini T, Legal S, Sorroche P, Galich AM, Pennisi P, Domene H, Jasper H (2000) Effect of deflazacort versus methylprednisone on growth, body composition, lipid profile, and bone mass after renal transplantation. The Deflazacort Study Group. Pediatr Nephrol 14: 682–688. 10.1007/s004670000337
    1. Finsterer J, Stollberger C (2003) The heart in human dystrophinopathies. Cardiology 99: 1–19. 10.1159/000068446
    1. Fiorillo AA, Tully CB, Damsker JM, Nagaraju K, Hoffman EP, Heier CR (2018) Muscle miRNAome shows suppression of chronic inflammatory miRNAs with both prednisone and vamorolone. Physiol Genomics 50: 735–745. 10.1152/physiolgenomics.00134.2017
    1. Flanigan KM, Ceco E, Lamar KM, Kaminoh Y, Dunn DM, Mendell JR, King WM, Pestronk A, Florence JM, Mathews KD, et al. (2013) LTBP4 genotype predicts age of ambulatory loss in Duchenne muscular dystrophy. Ann Neurol 73: 481–488. 10.1002/ana.23819
    1. Fraccarollo D, Berger S, Galuppo P, Kneitz S, Hein L, Schutz G, Frantz S, Ertl G, Bauersachs J (2011) Deletion of cardiomyocyte mineralocorticoid receptor ameliorates adverse remodeling after myocardial infarction. Circulation 123: 400–408. 10.1161/circulationaha.110.983023
    1. Funder JW, Pearce PT, Smith R, Smith AI (1988) Mineralocorticoid action: Target tissue specificity is enzyme, not receptor, mediated. Science 242: 583–585. 10.1126/science.2845584
    1. Griggs RC, Herr BE, Reha A, Elfring G, Atkinson L, Cwik V, McColl E, Tawil R, Pandya S, McDermott MP, et al. (2013) Corticosteroids in Duchenne muscular dystrophy: Major variations in practice. Muscle Nerve 48: 27–31. 10.1002/mus.23831
    1. Griggs RC, Miller JP, Greenberg CR, Fehlings DL, Pestronk A, Mendell JR, Moxley RT 3rd, King W, Kissel JT, Cwik V, et al. (2016) Efficacy and safety of deflazacort vs prednisone and placebo for Duchenne muscular dystrophy. Neurology 87: 2123–2131. 10.1212/wnl.0000000000003217
    1. Grossmann C, Scholz T, Rochel M, Bumke-Vogt C, Oelkers W, Pfeiffer AF, Diederich S, Bahr V (2004) Transactivation via the human glucocorticoid and mineralocorticoid receptor by therapeutically used steroids in CV-1 cells: A comparison of their glucocorticoid and mineralocorticoid properties. Eur J Endocrinol 151: 397–406. 10.1530/eje.0.1510397
    1. Grosso S, Farnetani M, Mostardini R, Cordelli D, Berardi R, Balestri P (2008) A comparative study of hydrocortisone versus deflazacort in drug-resistant epilepsy of childhood. Epilepsy Res 81: 80–85. 10.1016/j.eplepsyres.2008.04.016
    1. Guerron AD, Rawat R, Sali A, Spurney CF, Pistilli E, Cha HJ, Pandey GS, Gernapudi R, Francia D, Farajian V, et al. (2010) Functional and molecular effects of arginine butyrate and prednisone on muscle and heart in the mdx mouse model of Duchenne Muscular Dystrophy. PLoS One 5: e11220 10.1371/annotation/a82245dd-7379-4067-bd84-b0fc5ffb5869
    1. Hahn BH, Pletscher LS, Muniain M (1981) Immunosuppressive effects of deflazacort: A new glucocorticoid with bone-sparing and carbohydrate-sparing properties: Comparison with prednisone. J Rheumatol 8: 783–790
    1. Hammer S, Spika I, Sippl W, Jessen G, Kleuser B, Holtje HD, Schafer-Korting M (2003) Glucocorticoid receptor interactions with glucocorticoids: Evaluation by molecular modeling and functional analysis of glucocorticoid receptor mutants. Steroids 68: 329–339. 10.1016/s0039-128x(03)00030-8
    1. He BJ, Joiner ML, Singh MV, Luczak ED, Swaminathan PD, Koval OM, Kutschke W, Allamargot C, Yang J, Guan X, et al. (2011) Oxidation of CaMKII determines the cardiotoxic effects of aldosterone. Nat Med 17: 1610–1618. 10.1038/nm.2506
    1. Heier CR, Damsker JM, Yu Q, Dillingham BC, Huynh T, Van der Meulen JH, Sali A, Miller BK, Phadke A, Scheffer L, et al. (2013) VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects. EMBO Mol Med 5: 1569–1585. 10.1002/emmm.201302621
    1. Heydemann A, Ceco E, Lim JE, Hadhazy M, Ryder P, Moran JL, Beier DR, Palmer AA, McNally EM (2009) Latent TGF-beta-binding protein 4 modifies muscular dystrophy in mice. J Clin Invest 119: 3703–3712. 10.1172/jci39845
    1. Hiscott J, Marois J, Garoufalis J, D'Addario M, Roulston A, Kwan I, Pepin N, Lacoste J, Nguyen H, Bensi G, et al. (1993) Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: Evidence for a positive autoregulatory loop. Mol Cell Biol 13: 6231–6240. 10.1128/mcb.13.10.6231
    1. Hoffman EP, Brown RH Jr., Kunkel LM (1987) Dystrophin: The protein product of the Duchenne muscular dystrophy locus. Cell 51: 919–928. 10.1016/0092-8674(87)90579-4
    1. Hoffman EP, Riddle V, Siegler MA, Dickerson D, Backonja M, Kramer WG, Nagaraju K, Gordish-Dressman H, Damsker JM, McCall JM (2018) Phase 1 trial of vamorolone, a first-in-class steroid, shows improvements in side effects via biomarkers bridged to clinical outcomes. Steroids 134: 43–52. 10.1016/j.steroids.2018.02.010
    1. Hor KN, Taylor MD, Al-Khalidi HR, Cripe LH, Raman SV, Jefferies JL, O'Donnell R, Benson DW, Mazur W (2013) Prevalence and distribution of late gadolinium enhancement in a large population of patients with Duchenne muscular dystrophy: Effect of age and left ventricular systolic function. J Cardiovasc Magn Reson 15: 107 10.1186/1532-429x-15-107
    1. Investigators TR. (1996) Effectiveness of spironolactone added to an angiotensin-converting enzyme inhibitor and a loop diuretic for severe chronic congestive heart failure (the Randomized Aldactone Evaluation Study [RALES]). Am J Cardiol 78: 902–907. 10.1016/S0002-9149(96)00465-1
    1. Iqbal J, Andrew R, Cruden NL, Kenyon CJ, Hughes KA, Newby DE, Hadoke PW, Walker BR (2014) Displacement of cortisol from human heart by acute administration of a mineralocorticoid receptor antagonist. J Clin Endocrinol Metab 99: 915–922. 10.1210/jc.2013-2049
    1. Janssen PM, Murray JD, Schill KE, Rastogi N, Schultz EJ, Tran T, Raman SV, Rafael-Fortney JA (2014) Prednisolone attenuates improvement of cardiac and skeletal contractile function and histopathology by lisinopril and spironolactone in the mdx mouse model of Duchenne muscular dystrophy. PLoS One 9: e88360 10.1371/journal.pone.0088360
    1. Koenig M, Hoffman EP, Bertelson CJ, Monaco AP, Feener C, Kunkel LM (1987) Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals. Cell 50: 509–517. 10.1016/0092-8674(87)90504-6
    1. Libermann TA, Baltimore D (1990) Activation of interleukin-6 gene expression through the NF-kappa B transcription factor. Mol Cell Biol 10: 2327–2334. 10.1128/mcb.10.5.2327
    1. Lind U, Greenidge P, Gillner M, Koehler KF, Wright A, Carlstedt-Duke J (2000) Functional probing of the human glucocorticoid receptor steroid-interacting surface by site-directed mutagenesis. Gln-642 plays an important role in steroid recognition and binding. J Biol Chem 275: 19041–19049. 10.1074/jbc.m000228200
    1. Lother A, Berger S, Gilsbach R, Rosner S, Ecke A, Barreto F, Bauersachs J, Schutz G, Hein L (2011) Ablation of mineralocorticoid receptors in myocytes but not in fibroblasts preserves cardiac function. Hypertension 57: 746–754. 10.1161/hypertensionaha.110.163287
    1. Lowe J, Floyd KT, Rastogi N, Schultz EJ, Chadwick JA, Swager SA, Zins JG, Kadakia FK, Smart S, Gomez-Sanchez EP, et al. (2016) Similar efficacy from specific and non-specific mineralocorticoid receptor antagonist treatment of muscular dystrophy mice. J Neuromuscul Dis 3: 395–404. 10.3233/jnd-160173
    1. Mesa LE, Dubrovsky AL, Corderi J, Marco P, Flores D (1991) Steroids in Duchenne muscular dystrophy--deflazacort trial. Neuromuscul Disord 1: 261–266. 10.1016/0960-8966(91)90099-e
    1. Monaco AP, Neve RL, Colletti-Feener C, Bertelson CJ, Kurnit DM, Kunkel LM (1986) Isolation of candidate cDNAs for portions of the Duchenne muscular dystrophy gene. Nature 323: 646–650. 10.1038/323646a0
    1. Nigro G, Comi LI, Politano L, Bain RJ (1990) The incidence and evolution of cardiomyopathy in Duchenne muscular dystrophy. Int J Cardiol 26: 271–277. 10.1016/0167-5273(90)90082-g
    1. Peters J, Schluter T, Riegel T, Peters BS, Beineke A, Maschke U, Hosten N, Mullins JJ, Rettig R (2009) Lack of cardiac fibrosis in a new model of high prorenin hyperaldosteronism. Am J Physiol Heart Circ Physiol 297: H1845–H1852. 10.1152/ajpheart.01135.2008
    1. Peterson JM, Wang DJ, Shettigar V, Roof SR, Canan BD, Bakkar N, Shintaku J, Gu JM, Little SC, Ratnam NM, et al. (2018) NF-kappaB inhibition rescues cardiac function by remodeling calcium genes in a Duchenne muscular dystrophy model. Nat Commun 9: 3431 10.1038/s41467-018-05910-1
    1. Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, Bittman R, Hurley S, Kleiman J, Gatlin M (2003) Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med 348: 1309–1321. 10.1056/nejmoa030207
    1. Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, Palensky J, Wittes J (1999) The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 341: 709–717. 10.1056/nejm199909023411001
    1. Rafael-Fortney JA, Chimanji NS, Schill KE, Martin CD, Murray JD, Ganguly R, Stangland JE, Tran T, Xu Y, Canan BD, et al. (2011) Early treatment with lisinopril and spironolactone preserves cardiac and skeletal muscle in Duchenne muscular dystrophy mice. Circulation 124: 582–588. 10.1161/circulationaha.111.031716
    1. Rafestin-Oblin ME, Fagart J, Souque A, Seguin C, Bens M, Vandewalle A (2002) 11beta-hydroxyprogesterone acts as a mineralocorticoid agonist in stimulating Na+ absorption in mammalian principal cortical collecting duct cells. Mol Pharmacol 62: 1306–1313. 10.1124/mol.62.6.1306
    1. Raman SV, Hor KN, Mazur W, Halnon NJ, Kissel JT, He X, Tran T, Smart S, McCarthy B, Taylor MD, et al. (2015) Eplerenone for early cardiomyopathy in Duchenne muscular dystrophy: A randomised, double-blind, placebo-controlled trial. Lancet Neurol 14: 153–161. 10.1016/s1474-4422(14)70318-7
    1. Raman SV, Hor KN, Mazur W, He X, Kissel JT, Smart S, McCarthy B, Roble SL, Cripe LH (2017) Eplerenone for early cardiomyopathy in Duchenne muscular dystrophy: Results of a two-year open-label extension trial. Orphanet J Rare Dis 12: 39 10.1186/s13023-017-0590-8
    1. Ratman D, Vanden Berghe W, Dejager L, Libert C, Tavernier J, Beck IM, De Bosscher K (2013) How glucocorticoid receptors modulate the activity of other transcription factors: A scope beyond tethering. Mol Cell Endocrinol 380: 41–54. 10.1016/j.mce.2012.12.014
    1. Rickard AJ, Morgan J, Chrissobolis S, Miller AA, Sobey CG, Young MJ (2014) Endothelial cell mineralocorticoid receptors regulate deoxycorticosterone/salt-mediated cardiac remodeling and vascular reactivity but not blood pressure. Hypertension 63: 1033–1040. 10.1161/hypertensionaha.113.01803
    1. Rickard AJ, Morgan J, Tesch G, Funder JW, Fuller PJ, Young MJ (2009) Deletion of mineralocorticoid receptors from macrophages protects against deoxycorticosterone/salt-induced cardiac fibrosis and increased blood pressure. Hypertension 54: 537–543. 10.1161/hypertensionaha.109.131110
    1. Robinson CM, Hale PT, Carlin JM (2006) NF-kappa B activation contributes to indoleamine dioxygenase transcriptional synergy induced by IFN-gamma and tumor necrosis factor-alpha. Cytokine 35: 53–61. 10.1016/j.cyto.2006.07.007
    1. Rossignol P, Menard J, Fay R, Gustafsson F, Pitt B, Zannad F (2011) Eplerenone survival benefits in heart failure patients post-myocardial infarction are independent from its diuretic and potassium-sparing effects. Insights from an EPHESUS (Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study) substudy. J Am Coll Cardiol 58: 1958–1966. 10.1016/j.jacc.2011.04.049
    1. Ruhs S, Stratz N, Schlor K, Meinel S, Mildenberger S, Rabe S, Gekle M, Grossmann C (2012) Modulation of transcriptional mineralocorticoid receptor activity by nitrosative stress. Free Radic Biol Med 53: 1088–1100. 10.1016/j.freeradbiomed.2012.06.028
    1. Sanders B, Cummings EE (2017) Letter to Jeffrey S. Aronin, Chairman and Chief Executive Officer, Marathon Pharmaceuticals, LLC. Congress of the United States ()
    1. Silversides CK, Webb GD, Harris VA, Biggar DW (2003) Effects of deflazacort on left ventricular function in patients with Duchenne muscular dystrophy. Am J Cardiol 91: 769–772. 10.1016/s0002-9149(02)03429-x
    1. Smith WG. (1962) Spironolactone and gynaecomastia. Lancet 280: 886 10.1016/s0140-6736(62)90668-2
    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. (2009) Preclinical drug trials in the mdx mouse: Assessment of reliable and sensitive outcome measures. Muscle Nerve 39: 591–602. 10.1002/mus.21211
    1. Spurney CF, Guerron AD, Yu Q, Sali A, van der Meulen JH, Hoffman EP, Nagaraju K (2011) Membrane sealant Poloxamer P188 protects against isoproterenol induced cardiomyopathy in dystrophin deficient mice. BMC Cardiovasc Disord 11: 20 10.1186/1471-2261-11-20
    1. Sreetama SC, Chandra G, Van der Meulen JH, Ahmad MM, Suzuki P, Bhuvanendran S, Nagaraju K, Hoffman EP, Jaiswal JK (2018) Membrane stabilization by modified steroid offers a potential therapy for muscular dystrophy due to dysferlin deficit. Mol Ther 26: 2231–2242. 10.1016/j.ymthe.2018.07.021
    1. Sun Y, Zhang J, Lu L, Chen SS, Quinn MT, Weber KT (2002) Aldosterone-induced inflammation in the rat heart: Role of oxidative stress. Am J Pathol 161: 1773–1781. 10.1016/s0002-9440(10)64454-9
    1. Townsend D, Blankinship MJ, Allen JM, Gregorevic P, Chamberlain JS, Metzger JM (2007) Systemic administration of micro-dystrophin restores cardiac geometry and prevents dobutamine-induced cardiac pump failure. Mol Ther 15: 1086–1092. 10.1038/sj.mt.6300144
    1. Uaesoontrachoon K, Quinn JL, Tatem KS, Van Der Meulen JH, Yu Q, Phadke A, Miller BK, Gordish-Dressman H, Ongini E, Miglietta D, et al. (2014) Long-term treatment with naproxcinod significantly improves skeletal and cardiac disease phenotype in the mdx mouse model of dystrophy. Hum Mol Genet 23: 3239–3249. 10.1093/hmg/ddu033
    1. Ueda A, Okuda K, Ohno S, Shirai A, Igarashi T, Matsunaga K, Fukushima J, Kawamoto S, Ishigatsubo Y, Okubo T (1994) NF-kappa B and Sp1 regulate transcription of the human monocyte chemoattractant protein-1 gene. J Immunol 153: 2052–2063
    1. Wollert KC, Drexler H (2001) The role of interleukin-6 in the failing heart. Heart Fail Rev 6: 95–103. 10.1023/A:1011401825680
    1. Yang C, Balsells J, Chu HD, Cox JM, Crespo A, Ma X, Contino L, Brown P, Gao S, Zamlynny B, et al. (2015) Discovery of benzimidazole oxazolidinediones as novel and selective nonsteroidal mineralocorticoid receptor antagonists. ACS Med Chem Lett 6: 461–465. 10.1021/acsmedchemlett.5b00010
    1. Yasuda S, Townsend D, Michele DE, Favre EG, Day SM, Metzger JM (2005) Dystrophic heart failure blocked by membrane sealant poloxamer. Nature 436: 1025–1029. 10.1038/nature03844
    1. Young MJ, Rickard AJ (2015) Mineralocorticoid receptors in the heart: Lessons from cell-selective transgenic animals. J Endocrinol 224: R1–R13. 10.1530/joe-14-0471
    1. Zannad F, McMurray JJ, Krum H, van Veldhuisen DJ, Swedberg K, Shi H, Vincent J, Pocock SJ, Pitt B (2011) Eplerenone in patients with systolic heart failure and mild symptoms. N Engl J Med 364: 11–21. 10.1056/nejmoa1009492

Source: PubMed

3
Předplatit