Andrographolide attenuates skeletal muscle dystrophy in mdx mice and increases efficiency of cell therapy by reducing fibrosis

Daniel Cabrera, Jaime Gutiérrez, Claudio Cabello-Verrugio, Maria Gabriela Morales, Sergio Mezzano, Ricardo Fadic, Juan Carlos Casar, Juan L Hancke, Enrique Brandan, Daniel Cabrera, Jaime Gutiérrez, Claudio Cabello-Verrugio, Maria Gabriela Morales, Sergio Mezzano, Ricardo Fadic, Juan Carlos Casar, Juan L Hancke, Enrique Brandan

Abstract

Background: Duchenne muscular dystrophy (DMD) is characterized by the absence of the cytoskeletal protein dystrophin, muscle wasting, increased transforming growth factor type beta (TGF-β) signaling, and fibrosis. At the present time, the only clinically validated treatments for DMD are glucocorticoids. These drugs prolong muscle strength and ambulation of patients for a short term only and have severe adverse effects. Andrographolide, a bicyclic diterpenoid lactone, has traditionally been used for the treatment of colds, fever, laryngitis, and other infections with no or minimal side effects. We determined whether andrographolide treatment of mdx mice, an animal model for DMD, affects muscle damage, physiology, fibrosis, and efficiency of cell therapy.

Methods: mdx mice were treated with andrographolide for three months and skeletal muscle histology, creatine kinase activity, and permeability of muscle fibers were evaluated. Fibrosis and TGF-β signaling were evaluated by indirect immunofluorescence and Western blot analyses. Muscle strength was determined in isolated skeletal muscles and by a running test. Efficiency of cell therapy was determined by grafting isolated skeletal muscle satellite cells onto the tibialis anterior of mdx mice.

Results: mdx mice treated with andrographolide exhibited less severe muscular dystrophy than untreated dystrophic mice. They performed better in an exercise endurance test and had improved muscle strength in isolated muscles, reduced skeletal muscle impairment, diminished fibrosis and a significant reduction in TGF-β signaling. Moreover, andrographolide treatment of mdx mice improved grafting efficiency upon intramuscular injection of dystrophin-positive satellite cells.

Conclusions: These results suggest that andrographolide could be used to improve quality of life in individuals with DMD.

Keywords: Andrographolide; Cell therapy; DMD; Fibrosis; Skeletal muscle; mdx.

Figures

Figure 1
Figure 1
Andrographolide reduces skeletal muscle damage in mdx mice. To augment the extent of muscle fibrosis, three-month-old mdx mice were subjected to an exercise protocol for three months [6,25]. During this period, mice were treated with 1 mg/kg andrographolide or vehicle (intraperitoneal (ip) injections three times per week, six animals per group). (A) H&E staining of tibialis anterior (TA) muscles showed a striking reduction in the damaged areas of muscle in andrographolide-treated mdx mice compared with untreated mdx mice. Upper panel shows 100X magnification pictures (scale bars = 200 μm), while the bottom panel shows 400X magnification pictures (scale bars = 50 μm). (B) Evans blue dye uptake in TA muscle fibers from wild type (WT), vehicle-treated mdx mice, and andrographolide-treated mdx mice. Nuclei were labeled with Hoechst. Mice were injected ip with 1% Evans blue dye 24 hours before muscle fixation (scale bar = 200 μm). (C) Serum creatine kinase (CK) was measured to evaluate skeletal muscle damage. The bar graph shows a significant reduction in serum CK activity in andrographolide-treated mdx mice compared with vehicle-treated mdx mice. Values are expressed as mean ± SD of three independent experiments, using five mice for each experimental condition. (*P < 0.05 relative to WT mice; #P < 0.05 relative to vehicle-treated mdx mice). The recovery score was 49.5%.
Figure 2
Figure 2
Andrographolide reduces skeletal muscle fibrosis in mdx mice. Fibrosis was augmented as explained in the legend of Figure 1. During this period, mice were treated with 1 mg/kg andrographolide or vehicle (intraperitoneal (ip) injections three times per week, six animals per group). Indirect immunofluorescence analysis of (A) collagen I (green) and (B) fibronectin (red) in cryosections of tibialis anterior (TA) muscles from wild type (WT), vehicle-treated mdx mice, and andrographolide-treated mdx mice. Nuclei are stained in blue (Hoechst). Bar corresponds to 200 and 50 μm for 100X and 400X magnification pictures respectively.
Figure 3
Figure 3
Andrographolide reduces the amount of fibrotic proteins in mdx skeletal muscle. Experiments were performed as explained in the legend of Figure 2. Muscle extracts were obtained under each experimental condition and fibrotic proteins were analyzed by western blotting. (A) Fibronectin (FN) and (B) collagen type III (Col III) from tibialis anterior (TA) muscles of wild type (WT), vehicle-treated mdx mice, and andrographolide-treated mdx mice. GAPDH protein levels are shown as loading control; molecular weight markers are shown in kDa. Values are expressed as mean ± SD of three independent experiments, using three mice for each experimental condition. (*P < 0.05 relative to WT mice; #P < 0.05 relative to vehicle-treated mdx mice).
Figure 4
Figure 4
Andrographolide decreases NF-κB activity in vivo. Fibrosis was augmented as explained in the legend of Figure 1. During this period, mice were treated with 1 mg/kg andrographolide or vehicle (intraperitoneal (ip) injections three times per week, six animals per group). The activity of NF-κB was evaluated in formalin-fixed muscle samples by Southwestern blot analysis. A specific DNA was used to detect active NF-κB by immunofluorescence (red). The nuclei were stained with Hoechst (blue). (A) The upper panel shows merged red (active NF-κB) and blue (total nuclei) signals. Bar corresponds to 50 μm. (B) Quantification of total nuclei per field (in 400X magnification pictures).
Figure 5
Figure 5
Andrographolide reduces expression of pro-fibrotic factors and collagen type I in mdx mice. Fibrosis was augmented as explained in the legend of Figure 1. During this period, mice were treated with 1 mg/kg andrographolide or vehicle (intraperitoneal (ip) injections three times per week, four animals per group). (A) Transforming growth factor type beta (TGF-β1), (B) Connective tissue growth factor (CTGF) and (C) collagen type I mRNA levels were determined in tibialis anterior (TA) muscle from wild type (WT), vehicle-treated mdx mice, and andrographolide-treated mdx mice by qPCR using GAPDH as a reference gene. Values correspond to the mean dCT value ± SD of three independent experiments, normalized to WT levels (*P < 0.05 relative to WT mice; #P < 0.05 relative to vehicle-treated mdx mice). The recovery scores for TGF-β1 and CTGF were 56.3% and 63.2 respectively.
Figure 6
Figure 6
Andrographolide reduces transforming growth factor type beta (TGF-β) signaling pathway activity in mdx mice. Indirect immunofluorescence analysis of (A) p-Smad-2 and (D) p-Smad-3 (intracellular TGF-β1 mediators) in cryosections of tibialis anterior (TA) muscle from vehicle-treated and andrographolide-treated mdx mice. Bar corresponds to 200 μm. (B,E) Ratio of p-Smad-2 and p-Smad-3 respectively versus total nuclei. (C,F) Quantification of total nuclei per field (in 400X magnification pictures). Values are expressed as mean ± SD of three independent experiments, using three mice for each experimental condition.
Figure 7
Figure 7
Andrographolide increases skeletal muscle strength in mdx mice. Fibrosis was augmented as explained in the legend of Figure 1. During this period, mice were treated with 1 mg/kg andrographolide or vehicle (intraperitoneal (ip) injections three times per week, six animals per group). (A) Tibialis anterior (TA) muscles were isolated from wild type (WT), vehicle-treated mdx mice, and andrographolide-treated mdx mice to evaluate isometric specific force (mN/mm2) at different stimulation frequencies (pulses per second, pps). (B) Bar graph shows tetanic specific force. Values are represented as percentage of specific isometric force generated by WT muscle (*P < 0.05 relative to WT mice; #P < 0.05 relative to vehicle-treated mdx mice). The recovery score for the twitch force measurement was 54.2%. (C) Bar graph showing twitch force (*P < 0.05 relative to WT mice; #P < 0.05 relative to vehicle-treated mdx mice). The recovery score for the tetanus measurement was 50.3%. (D) Mice were subjected to an exercise challenge on the treadmill at 15 meters/minute for five minutes and the number of set-backs was counted (n = 7, *P < 0.05 relative to WT mice; #P < 0.05 relative to vehicle-treated mdx mice). The recovery score for this measurement was 45.5%.
Figure 8
Figure 8
Muscle stem cell therapy with satellite cells (SC) is improved by the reduction in muscle fibrosis following treatment with andrographolide. (A) Fibrosis was augmented as explained in the legend of Figure 1. During this period, mice were treated with andrographolide or vehicle (intraperitoneal (ip) injections three times per week, six animals per group). One week after the last drug administration (seven-month-old mice), 500 freshly isolated satellite cells purified from wild type (WT) mice were transplanted into both tibialis anterior (TA) muscles of each mdx mouse. At four weeks after engraftment, the number of fibers expressing dystrophin and collagen I was determined by immunofluorescence analysis of cryosections. The images are representative of two experimental groups with six mice per group. Bar corresponds to 200 μm. (B) Quantification of the data obtained in (A) showing the number of myofibers expressing dystrophin per TA muscle in each case. (C) The number of satellite cells (PAX-7-positive nuclei) was determined for isolated single muscle fibers from extensor digitorum longus muscle (EDL) of each case as an indicator of endogenous SC survival. (D) 500 WT SC freshly isolated as in (A), from the C57-EGFP mice were engrafted in mice treated as in (A) (six animals per group). Both TA muscles were dissected immediately after engraftment or after 2 or 15 days from two animals in each case. Total genomic DNA was purified. EGFP transgene present in the engrafted muscle was detected by qPCR. Values are expressed as mean ± SD of three independent experiments. (*P<0.05 relative to vehicle-treated mice, **P<0.01 relative to vehicle-treated mice at day 15).

References

    1. Kapsa R, Kornberg AJ, Byrne E. Novel therapies for Duchenne muscular dystrophy. Lancet Neurol. 2003;2:299–310. doi: 10.1016/S1474-4422(03)00382-X.
    1. Blau HM, Webster C, Pavlath GK. Defective myoblasts identified in Duchenne muscular dystrophy. Proc Natl Acad Sci USA. 1983;80:4856–4860. doi: 10.1073/pnas.80.15.4856.
    1. Allen DG, Whitehead NP. Duchenne muscular dystrophy - what causes the increased membrane permeability in skeletal muscle? Int J Biochem Cell Biol. 2011;43:290–294. doi: 10.1016/j.biocel.2010.11.005.
    1. Alvarez K, Fadic R, Brandan E. Augmented synthesis and differential localization of heparan sulfate proteoglycans in Duchenne muscular dystrophy. J Cell Biochem. 2002;85:703–713. doi: 10.1002/jcb.10184.
    1. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214:199–210. doi: 10.1002/path.2277.
    1. Morales MG, Gutierrez J, Cabello-Verrugio C, Cabrera D, Lipson KE, Goldschmeding R, Brandan E. Reducing CTGF/CCN2 slows mdx muscle dystrophy and improves cell-therapy. Hum Mol Genet. 2013;22:4938–4951. doi: 10.1093/hmg/ddt352.
    1. Brandan E, Gutierrez J. Role of proteoglycans in the regulation of the skeletal muscle fibrotic response. FEBS J. 2013;280:4109–4117. doi: 10.1111/febs.12278.
    1. Varga J, Brenner DA, Phan SH. Fibrosis Research: Methods and Protocols. Totowa, NJ: Humana Press; 2005. p. 392.
    1. Zhou L, Lu H. Targeting fibrosis in Duchenne muscular dystrophy. J Neuropathol Exp Neurol. 2010;69:771–776. doi: 10.1097/NEN.0b013e3181e9a34b.
    1. Desguerre I, Mayer M, Leturcq F, Barbet JP, Gherardi RK, Christov C. Endomysial fibrosis in Duchenne muscular dystrophy: a marker of poor outcome associated with macrophage alternative activation. J Neuropathol Exp Neurol. 2009;68:762–773. doi: 10.1097/NEN.0b013e3181aa31c2.
    1. Shen YC, Chen CF, Chiou WF. Andrographolide prevents oxygen radical production by human neutrophils: possible mechanism(s) involved in its anti-inflammatory effect. Br J Pharmacol. 2002;135:399–406. doi: 10.1038/sj.bjp.0704493.
    1. Rajagopal S, Kumar RA, Deevi DS, Satyanarayana C, Rajagopalan R. Andrographolide, a potential cancer therapeutic agent isolated from Andrographis paniculata. J Exp Ther Oncol. 2003;3:147–158. doi: 10.1046/j.1359-4117.2003.01090.x.
    1. Calabrese C, Berman SH, Babish JG, Ma X, Shinto L, Dorr M, Wells K, Wenner CA, Standish LJ. A phase I trial of andrographolide in HIV positive patients and normal volunteers. Phytother Res. 2000;14:333–338. doi: 10.1002/1099-1573(200008)14:5<333::AID-PTR584>;2-D.
    1. Lee TY, Lee KC, Chang HH. Modulation of the cannabinoid receptors by andrographolide attenuates hepatic apoptosis following bile duct ligation in rats with fibrosis. Apoptosis. 2010;15:904–914. doi: 10.1007/s10495-010-0502-z.
    1. Lee MJ, Rao YK, Chen K, Lee YC, Chung YS, Tzeng YM. Andrographolide and 14-deoxy-11,12-didehydroandrographolide from Andrographis paniculata attenuate high glucose-induced fibrosis and apoptosis in murine renal mesangial cell lines. J Ethnopharmacol. 2010;132:497–505. doi: 10.1016/j.jep.2010.07.057.
    1. Ye JF, Zhu H, Zhou ZF, Xiong RB, Wang XW, Su LX, Luo BD. Protective mechanism of andrographolide against carbon tetrachloride-induced acute liver injury in mice. Biol Pharm Bull. 2011;34:1666–1670. doi: 10.1248/bpb.34.1666.
    1. Xia YF, Ye BQ, Li YD, Wang JG, He XJ, Lin X, Yao X, Ma D, Slungaard A, Hebbel RP, Key NS, Geng JG. Andrographolide attenuates inflammation by inhibition of NF-kappa B activation through covalent modification of reduced cysteine 62 of p50. J Immunol. 2004;173:4207–4217. doi: 10.4049/jimmunol.173.6.4207.
    1. Acharyya S, Villalta SA, Bakkar N, Bupha-Intr T, Janssen PM, Carathers M, Li ZW, Beg AA, Ghosh S, Sahenk Z, Weinstein M, Gardner KL, Rafael-Fortney JA, Karin M, Tidball JG, Baldwin AS, Guttridge DC. Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy. J Clin Invest. 2007;117:889–901. doi: 10.1172/JCI30556.
    1. Peterson JM, Guttridge DC. Skeletal muscle diseases, inflammation, and NF-kappaB signaling: insights and opportunities for therapeutic intervention. Int Rev Immunol. 2008;27:375–387. doi: 10.1080/08830180802302389.
    1. Peterson JM, Kline W, Canan BD, Ricca DJ, Kaspar B, Delfín DA, DiRienzo K, Clemens PR, Robbins PD, Baldwin AS, Flood P, Kaumaya P, Freitas M, Kornegay JN, Mendell JR, Rafael-Fortney JA, Guttridge DC, Janssen PM. 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. Gargioli C, Coletta M, De Grandis F, Cannata SM, Cossu G. PlGF-MMP-9-expressing cells restore microcirculation and efficacy of cell therapy in aged dystrophic muscle. Nat Med. 2008;14:973–978. doi: 10.1038/nm.1852.
    1. Rowland LP. Pathogenesis of muscular dystrophies. Arch Neurol. 1976;33:315–321. doi: 10.1001/archneur.1976.00500050001001.
    1. De Luca A, Pierno S, Liantonio A, Cetrone M, Camerino C, Fraysse B, Mirabella M, Servidei S, Ruegg UT, Conte Camerino D. Enhanced dystrophic progression in mdx mice by exercise and beneficial effects of taurine and insulin-like growth factor-1. J Pharmacol Exp Ther. 2003;304:453–463. doi: 10.1124/jpet.102.041343.
    1. De Luca A, Nico B, Liantonio A, Didonna MP, Fraysse B, Pierno S, Burdi R, Mangieri D, Rolland JF, Camerino C, Zallone A, Confalonieri P, Andreetta F, Arnoldi E, Courdier-Fruh I, Magyar JP, Frigeri A, Pisoni M, Svelto M, Conte Camerino D. A multidisciplinary evaluation of the effectiveness of cyclosporine a in dystrophic mdx mice. Am J Pathol. 2005;166:477–489. doi: 10.1016/S0002-9440(10)62270-5.
    1. Cabello-Verrugio C, Morales MG, Cabrera D, Vio CP, Brandan E. Angiotensin II receptor type 1 blockade decreases CTGF/CCN2-mediated damage and fibrosis in normal and dystrophic skeletal muscles. J Cell Mol Med. 2012;16:752–764. doi: 10.1111/j.1582-4934.2011.01354.x.
    1. Morales MG, Cabello-Verrugio C, Santander C, Cabrera D, Goldschmeding R, Brandan E. CTGF/CCN-2 over-expression can directly induce features of skeletal muscle dystrophy. J Pathol. 2011;225:490–501. doi: 10.1002/path.2952.
    1. Grounds MD, Torrisi J. Anti-TNFalpha (Remicade) therapy protects dystrophic skeletal muscle from necrosis. FASEB J. 2004;18:676–682. doi: 10.1096/fj.03-1024com.
    1. Grounds MD, Radley HG, Lynch GS, Nagaraju K, De Luca A. Towards developing standard operating procedures for pre-clinical testing in the mdx mouse model of Duchenne muscular dystrophy. Neurobiol Dis. 2008;31:1–19. doi: 10.1016/j.nbd.2008.03.008.
    1. Straub V, Rafael JA, Chamberlain JS, Campbell KP. Animal models for muscular dystrophy show different patterns of sarcolemmal disruption. J Cell Biol. 1997;139:375–385. doi: 10.1083/jcb.139.2.375.
    1. Hernandez-Presa MA, Gomez-Guerrero C, Egido J. In situ non-radioactive detection of nuclear factors in paraffin sections by Southwestern histochemistry. Kidney Int. 1999;55:209–214. doi: 10.1046/j.1523-1755.1999.00226.x.
    1. Morales MG, Cabrera D, Cespedes C, Vio CP, Vazquez Y, Brandan E, Cabello-Verrugio C. Inhibition of the angiotensin-converting enzyme decreases skeletal muscle fibrosis in dystrophic mice by a diminution in the expression and activity of connective tissue growth factor (CTGF/CCN-2) Cell Tissue Res. 2013;353:173–187. doi: 10.1007/s00441-013-1642-6.
    1. Villalta SA, Rinaldi C, Deng B, Liu G, Fedor B, Tidball JG. Interleukin-10 reduces the pathology of mdx muscular dystrophy by deactivating M1 macrophages and modulating macrophage phenotype. Hum Mol Genet. 2011;20:790–805. doi: 10.1093/hmg/ddq523.
    1. Gregorevic P, Plant DR, Leeding KS, Bach LA, Lynch GS. Improved contractile function of the mdx dystrophic mouse diaphragm muscle after insulin-like growth factor-I administration. Am J Pathol. 2002;161:2263–2272. doi: 10.1016/S0002-9440(10)64502-6.
    1. Barton ER, Morris L, Kawana M, Bish LT, Toursel T. Systemic administration of L-arginine benefits mdx skeletal muscle function. Muscle Nerve. 2005;32:751–760. doi: 10.1002/mus.20425.
    1. Bogdanovich S, McNally EM, Khurana TS. Myostatin blockade improves function but not histopathology in a murine model of limb-girdle muscular dystrophy 2C. Muscle Nerve. 2008;37:308–316. doi: 10.1002/mus.20920.
    1. Collins CA, Olsen I, Zammit PS, Heslop L, Petrie A, Partridge TA, Morgan JE. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell. 2005;122:289–301. doi: 10.1016/j.cell.2005.05.010.
    1. Gillis JM. Multivariate evaluation of the functional recovery obtained by the overexpression of utrophin in skeletal muscles of the mdx mouse. Neuromuscul Disord. 2002;12(Suppl 1):S90–S94.
    1. Radley HG, Davies MJ, Grounds MD. Reduced muscle necrosis and long-term benefits in dystrophic mdx mice after cV1q (blockade of TNF) treatment. Neuromuscul Disord. 2008;18:227–238. doi: 10.1016/j.nmd.2007.11.002.
    1. Caceres S, Cuellar C, Casar JC, Garrido J, Schaefer L, Kresse H, Brandan E. Synthesis of proteoglycans is augmented in dystrophic mdx mouse skeletal muscle. Eur J Cell Biol. 2000;79:173–181. doi: 10.1078/S0171-9335(04)70020-5.
    1. Andreetta F, Bernasconi P, Baggi F, Ferro P, Oliva L, Arnoldi E, Cornelio F, Mantegazza R, Confalonieri P. Immunomodulation of TGF-beta 1 in mdx mouse inhibits connective tissue proliferation in diaphragm but increases inflammatory response: implications for antifibrotic therapy. J Neuroimmunol. 2006;175:77–86. doi: 10.1016/j.jneuroim.2006.03.005.
    1. Cohn RD, van Erp C, Habashi JP, Soleimani AA, Klein EC, Lisi MT, Gamradt M, ap Rhys CM, Holm TM, Loeys BL, Ramirez F, Judge DP, Ward CW, Dietz HC. Angiotensin II type 1 receptor blockade attenuates TGF-beta-induced failure of muscle regeneration in multiple myopathic states. Nat Med. 2007;13:204–210. doi: 10.1038/nm1536.
    1. Vial C, Zuniga LM, Cabello-Verrugio C, Canon P, Fadic R, Brandan E. Skeletal muscle cells express the profibrotic cytokine connective tissue growth factor (CTGF/CCN2), which induces their dedifferentiation. J Cell Physiol. 2008;215:410–421. doi: 10.1002/jcp.21324.
    1. De Luca A. Pre-clinical drug tests in the mdx mouse as a model of dystrophinopathies: an overview. Acta Myol. 2012;31:40–47.
    1. Nelson CA, Hunter RB, Quigley LA, Girgenrath S, Weber WD, McCullough JA, Dinardo CJ, Keefe KA, Ceci L, Clayton NP, McVie-Wylie A, Cheng SH, Leonard JP, Wentworth BM. Inhibiting TGF-beta activity improves respiratory function in mdx mice. Am J Pathol. 2011;178:2611–2621. doi: 10.1016/j.ajpath.2011.02.024.
    1. Huebner KD, Jassal DS, Halevy O, Pines M, Anderson JE. Functional resolution of fibrosis in mdx mouse dystrophic heart and skeletal muscle by halofuginone. Am J Physiol Heart Circ Physiol. 2008;294:H1550–H1561. doi: 10.1152/ajpheart.01253.2007.
    1. Turgeman T, Hagai Y, Huebner K, Jassal DS, Anderson JE, Genin O, Nagler A, Halevy O, Pines M. Prevention of muscle fibrosis and improvement in muscle performance in the mdx mouse by halofuginone. Neuromuscul Disord. 2008;18:857–868. doi: 10.1016/j.nmd.2008.06.386.
    1. Pines M, Halevy O. Halofuginone and muscular dystrophy. Histol Histopathol. 2011;26:135–146.
    1. MacDonald EM, Cohn RD. TGFbeta signaling: its role in fibrosis formation and myopathies. Curr Opin Rheumatol. 2012;24:628–634. doi: 10.1097/BOR.0b013e328358df34.
    1. Acuna MJ, Pessina P, Olguin H, Cabrera D, Vio CP, Bader M, Munoz-Canoves P, Santos RA, Cabello-Verrugio C, Brandan E. Restoration of muscle strength in dystrophic muscle by angiotensin-1-7 through inhibition of TGF-beta signalling. Hum Mol Genet. 2014;23:1237–1249. doi: 10.1093/hmg/ddt514.
    1. Wehling-Henricks M, Lee JJ, Tidball JG. Prednisolone decreases cellular adhesion molecules required for inflammatory cell infiltration in dystrophin-deficient skeletal muscle. Neuromuscul Disord. 2004;14:483–490. doi: 10.1016/j.nmd.2004.04.008.
    1. Hoffman EP, Reeves E, Damsker J, Nagaraju K, McCall JM, Connor EM, Bushby K. Novel approaches to corticosteroid treatment in Duchenne muscular dystrophy. Phys Med Rehabil Clin N Am. 2012;23:821–828. doi: 10.1016/j.pmr.2012.08.003.
    1. Wong BL, Christopher C. Corticosteroids in Duchenne muscular dystrophy: a reappraisal. J Child Neurol. 2002;17:183–190. doi: 10.1177/088307380201700306.
    1. Lim JC, Chan TK, Ng DS, Sagineedu SR, Stanslas J, Wong WS. Andrographolide and its analogues: versatile bioactive molecules for combating inflammation and cancer. Clin Exp Pharmacol Physiol. 2012;39:300–310. doi: 10.1111/j.1440-1681.2011.05633.x.
    1. Cabello-Verrugio C, Acuna MJ, Morales MG, Becerra A, Simon F, Brandan E. Fibrotic response induced by angiotensin-II requires NAD(P)H oxidase-induced reactive oxygen species (ROS) in skeletal muscle cells. Biochem Biophys Res Commun. 2011;410:665–670. doi: 10.1016/j.bbrc.2011.06.051.
    1. Mezzano V, Cabrera D, Vial C, Brandan E. Constitutively activated dystrophic muscle fibroblasts show a paradoxical response to TGF-beta and CTGF/CCN2. J Cell Commun Signal. 2007;1:205–217. doi: 10.1007/s12079-008-0018-2.
    1. Li G, Xie Q, Shi Y, Li D, Zhang M, Jiang S, Zhou H, Lu H, Jin Y. Inhibition of connective tissue growth factor by siRNA prevents liver fibrosis in rats. J Gene Med. 2006;8:889–900. doi: 10.1002/jgm.894.
    1. Guha M, Xu ZG, Tung D, Lanting L, Natarajan R. Specific down-regulation of connective tissue growth factor attenuates progression of nephropathy in mouse models of type 1 and type 2 diabetes. FASEB J. 2007;21:3355–3368. doi: 10.1096/fj.06-6713com.
    1. Klingler W, Jurkat-Rott K, Lehmann-Horn F, Schleip R. The role of fibrosis in Duchenne muscular dystrophy. Acta Myol. 2012;31:184–195.
    1. Leask A, Abraham DJ. TGF-beta signaling and the fibrotic response. FASEB J. 2004;18:816–827. doi: 10.1096/fj.03-1273rev.
    1. Ng CM, Cheng A, Myers LA, Martinez-Murillo F, Jie C, Bedja D, Gabrielson KL, Hausladen JM, Mecham RP, Judge DP, Dietz HC. TGF-beta-dependent pathogenesis of mitral valve prolapse in a mouse model of Marfan syndrome. J Clin Invest. 2004;114:1586–1592. doi: 10.1172/JCI200422715.
    1. Fukushima K, Badlani N, Usas A, Riano F, Fu F, Huard J. The use of an antifibrosis agent to improve muscle recovery after laceration. Am J Sports Med. 2001;29:394–402.
    1. de Winter P, Leoni P, Abraham D. Connective tissue growth factor: structure-function relationships of a mosaic, multifunctional protein. Growth Factors. 2008;26:80–91. doi: 10.1080/08977190802025602.
    1. Wang Q, Usinger W, Nichols B, Gray J, Xu L, Seeley TW, Brenner M, Guo G, Zhang W, Oliver N, Lin A, Yeowell D. Cooperative interaction of CTGF and TGF-beta in animal models of fibrotic disease. Fibrogenesis Tissue Repair. 2011;4:4. doi: 10.1186/1755-1536-4-4.
    1. Chaqour B, Yang R, Sha Q. Mechanical stretch modulates the promoter activity of the profibrotic factor CCN2 through increased actin polymerization and NF-kappaB activation. J Biol Chem. 2006;281:20608–20622. doi: 10.1074/jbc.M600214200.

Source: PubMed

3
Sottoscrivi