Tamoxifen prolongs survival and alleviates symptoms in mice with fatal X-linked myotubular myopathy

Elinam Gayi, Laurence A Neff, Xènia Massana Muñoz, Hesham M Ismail, Marta Sierra, Thomas Mercier, Laurent A Décosterd, Jocelyn Laporte, Belinda S Cowling, Olivier M Dorchies, Leonardo Scapozza, Elinam Gayi, Laurence A Neff, Xènia Massana Muñoz, Hesham M Ismail, Marta Sierra, Thomas Mercier, Laurent A Décosterd, Jocelyn Laporte, Belinda S Cowling, Olivier M Dorchies, Leonardo Scapozza

Abstract

X-linked myotubular myopathy (XLMTM, also known as XLCNM) is a severe congenital muscular disorder due to mutations in the myotubularin gene, MTM1. It is characterized by generalized hypotonia, leading to neonatal death of most patients. No specific treatment exists. Here, we show that tamoxifen, a well-known drug used against breast cancer, rescues the phenotype of Mtm1-deficient mice. Tamoxifen increases lifespan several-fold while improving overall motor function and preventing disease progression including lower limb paralysis. Tamoxifen corrects functional, histological and molecular hallmarks of XLMTM, with improved force output, myonuclei positioning, myofibrillar structure, triad number, and excitation-contraction coupling. Tamoxifen normalizes the expression level of the XLMTM disease modifiers DNM2 and PI3KC2B, likely contributing to the phenotypic rescue. Our findings demonstrate that tamoxifen is a promising candidate for clinical evaluation in XLMTM patients.

Conflict of interest statement

J.L. and B.S.C. are scientific advisors for Dynacure. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Tamoxifen rescues the phenotype of XLMTM mice. Life-long oral tamoxifen delays disease progression in XLMTM mice. Wild type (WT) and Mtm1−/y mice were fed control or tamoxifen (TAM)-supplemented diets from weaning onward. a Photographs of WT and Mtm1−/y mice, illustrating disease severity in young untreated mice and protection conferred by tamoxifen in adult and old mice. b Kaplan–Meier curves showing the effects of treatments on mouse survival. Early death of untreated Mtm1−/y mice contrasts with prolongation of life span in tamoxifen-treated littermates. Data are from 8 to 18 mice per group. *P ≤ 0.05; ****P ≤ 0.0001. Log-rank (Mantel–Cox) test. c Disease progression was assessed three times per week using a 5-grades clinical scale: muscle function was scored as 1 (normal function of hind limbs), 2 (difficulty in spreading toes), 3 (evident weakness in legs), 4 (paralysis of one hind limb), or 5 (complete paralysis of both legs). WT mice had a clinical score of 1 (normal) throughout the study. Untreated Mtm1−/y mice quickly reached a high clinical grade, whereas disease progressed much slower in tamoxifen-treated littermates. d Mouse motor function was assessed weekly via a horizontal grid-hanging test. The score of untreated Mtm1−/y mice declined quickly. Tamoxifen preserved motor function of Mtm1−/y mice close to WT values. e Mouse body weight was recorded 3 times per week. Tamoxifen affected the growth of WT but not of Mtm1−/y mice. Mtm1−/y mice remained smaller than WT mice throughout. be Symbols and TAM doses (0.3, 3, and 30 mg kg−1 of diet) are shown on the right-hand side. c, d, e Data represent the mean ± s.e.m. of 8 to 18 mice as defined in a. *P ≤ 0.05; **P ≤ 0.01; ****P ≤ 0.0001; ns non-significant. One-way ANOVA with Fisher’s least significance difference (LSD) post-test
Fig. 2
Fig. 2
Tamoxifen augments the strength of leg muscles of XLMTM mice. Electrically evoked triceps contractions were recorded under isometric conditions and contractile features analyzed in 42, 84, and 210-day-old mice fed placebo or tamoxifen-supplemented (30 mg kg−1) diets. a Average traces showing absolute phasic (twitch) contraction of the triceps at 42 days (D42). Tamoxifen significantly enhanced the force (tension) in Mtm1−/y mice. b Specific phasic force (see Online Methods for details) of D42 Mtm1−/y mice augmented with tamoxifen. c Specific phasic force of Mtm1−/y mice increased with tamoxifen at all examined ages. d Average traces showing triceps absolute force-frequency curves at D42. Tamoxifen more than doubled the tetanic force of Mtm1−/y mice. e Specific force of Mtm1−/y mice markedly augmented with tamoxifen at all stimulation frequencies. f Specific tetanic force of Mtm1−/y mice was considerably improved at all studied ages. In triceps examined after various treatment durations, tamoxifen rescued the impaired time to twitch peak of Mtm1−/y mice (g) but had no impact on the time required for half-relaxation from peak (h). D42 panels: WT wild type; legend in a stands for a, b, d, e; data represent the mean ± s.e.m. of n = 7 triceps per group. c, f, g, h: black triangles illustrate increasing age (42–84 and 210 days) within each treatment group; columns from left to right: data represent the mean ± s.e.m. of 7; 8; 7; 7; 7; 13; 7; 0; 0; 7; 8; 6 triceps, respectively. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001; ns non-significant, Ø no surviving mice. One-way ANOVA followed by Fisher’s LSD post-test
Fig. 3
Fig. 3
Tamoxifen mitigates muscle structure and improves sarcomeric organization in XLMTM mice. Muscle structure and sarcomere ultrastructure were examined by histology and transmission electron microscopy, respectively, in 42-day-old (D42) mice either untreated (control; Ctrl) or treated with tamoxifen (TAM; 30 mg kg−1 of diet). a Representative pictures of hematoxylin–eosin stained sections from the tibialis anterior (TA) of D42 mice. Note the small size of the Mtm1−/y myofibres and the presence of mislocalized nuclei, a hallmark of Mtm1−/y mice and XLMTM patients (arrows). b Representative pictures of succinate dehydrogenase (SDH) activity in TA sections of D42 mice, demonstrating abnormal distribution of oxidative staining. The intense staining forming a ring at the periphery of many Mtm1−/y myofibres (examples shown by asterisks) is due to accumulated mitochondria and other organelles and is a hallmark of the pathology. c The percentage of nuclei abnormally positioned (either internally or centrally located) in TA myofibres of D42 mice were counted from hematoxylin–eosin-stained sections. That feature was reduced by 53.3% with TAM. Data represent the mean ± s.e.m. of n = 2–4 TA per group. ****P ≤ 0.0001. One-way ANOVA followed by Fisher’s LSD post-test. d Sarcomere ultrastructure revealed by transmission electron microscopy in the TA from untreated (control) and tamoxifen-treated Mtm1−/y mice at D42. Note overall disorganization of the sarcomeres in untreated TA with disruption of the Z-lines (structures running perpendicular to the sarcomeres and holding myofibrils together; arrowheads) and shifted sarcomeres (dashed arrow). Tamoxifen ameliorated the organization of the sarcomeres as demonstrated by in-register Z-lines in adjacent myofibrils (arrowheads). The bar represents 50 µm in a and b, and 2 µm in d
Fig. 4
Fig. 4
Tamoxifen reduces the expression of XLMTM disease modifiers and alters the expression of estrogen receptors. The mRNA and protein levels of known XLMTM disease modifiers and of estrogen receptors, which mediate most tamoxifen actions, were determined in gastrocnemius muscle of 42-days old (D42) wild type (WT) and Mtm1−/y mice, either untreated (control; Ctrl) or tamoxifen (TAM)-treated (30 mg kg−1 of diet). a mRNA levels normalized to glyceraldehyde-3-phosphate dehydrogenase (Gapdh). From left to right, relative expression (as percentage of WT Ctrl) of mRNA encoding amphiphysin 2/BIN1 (Bin1; all transcripts), muscle-specific BIN1 (Bin1; exon 11-containing transcript), dynamin-2 (Dnm2), PI3KC2B (Pik3c2b), estrogen receptor (ER) α (Esr1), and ERβ (Esr2). b Representative immunoblots of proteins of interest, as indicated. DNM2: dynamin 2; MyHC: myosin heavy chains; GAPDH: glyceraldehyde-3-phosphate dehydrogenase. Position of molecular weight markers (kDa) is shown. cf Levels (normalized to GAPDH and expressed as percentage of WT Ctrl) of proteins selected for their role in XLMTM and tamoxifen signaling. c Proteins involved in the “MAD”-pathway. From top to bottom: myotubularin (MTM1), amphiphysin 2/BIN1 (pan-isoforms), amphiphysin 2/BIN1 (muscle-specific isoform), dynamin-2 (DNM2). d Other disease modifiers and protein deregulated in absence of MTM1. From top to bottom: desmin, dysferlin, PI3KC2B. e Estrogen receptors. Top: ERα; bottom: ERβ. f Myosin heavy chains, a major constituent of sarcomeres. Data represent the mean ± s.e.m. of n = 6–8 muscles per group. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001; ns non-significant. One-way ANOVA followed by Fisher’s LSD post-test
Fig. 5
Fig. 5
Tamoxifen improves triad density and excitation-contraction coupling, Triads and excitation–contraction coupling were examined in 42 days old (D42) mice either untreated (control; Ctrl) or treated with tamoxifen (TAM; 30 mg kg−1 in diet). a Triads—specialized membrane structures made of a t-tubule (tub) flanked by terminal cisternae (cis) arising from the sarcoplasmic reticulum and controlling Ca2+ release—were visualized by transmission electron microscopy (TEM) in the tibialis anterior (TA) of mice at D42. Note the abnormal shape of the remaining triads in the Mtm1−/y mouse and recovery with tamoxifen treatment. The bar represents 100 nm. b The number of well-positioned triads per sarcomere unit was determined from TEM pictures in TA from wild type (WT) and Mtm1−/y mice at D42. TAM partly rescued the much decreased triad density in Mtm1−/y mice. Mean ± s.e.m. of n = 2–3 mice. ce Excitation–contraction coupling assessed via live imaging of Ca2+ fluxes induced by KCl depolarization in single FDB myofibers (see Online Methods for details). c Average traces of the responses of FDB fibers to KCl, an experimental setting that mimics muscle depolarization (n = 17–29 fibers). d Representative images (pseudo-colored) illustrating cytosolic Ca2+ levels in FDB fibers at baseline (−; before KCl pulse) and at the peak of KCl-induced response (+). The bar represents 100 μm. e Quantification of cytosolic Ca2+ at the peak response. TAM enhanced Mtm1−/y cytosolic Ca2+ to levels found in WT. Data shown in e represent, from left to right, the mean ± s.e.m. of n = 29, 53, 53, and 31 fibers. f Representative western blots of DHPR and RyR1. g, h Quantification of DHPR and RyR1, respectively. Mean ± s.e.m. of n = 7 muscles. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001; ns non-significant. One-way ANOVA followed by Fisher’s LSD post-test
Fig. 6
Fig. 6
Myopathic features after long-term tamoxifen treatment, Muscle structure and triads were examined by histology and transmission electron microscopy, respectively, in wild type (WT) and Mtm1−/y mice during long-term treatment with tamoxifen (30 mg kg−1 in diet). a Representative pictures of tibialis anterior (TA) sections from untreated WT and Mtm1−/y mice at D42, and tamoxifen-treated Mtm1−/y mice at D42, D84, and D210 stained with hematoxylin–eosin (HE) or for succinate dehydrogenase (SDH) activity. Muscle structure of Mtm1−/y mice was stable over the duration of tamoxifen treatment. The bar represents 50 µm. b Scatter plots showing the distribution of TA myofiber diameter in mice at D42, D84, and D210 (black triangles illustrate increasing age). Fiber size remained almost constant from D42 to D210. N = 600 myofibers per group. c The percentage of abnormally positioned nuclei (either internally or centrally located) in TA myofibres of mice at D42, D84, and D210 were counted from hematoxylin–eosin-stained sections. The density of abnormally positioned nuclei increased significantly with ageing. Mean ± s.e.m. of n = 2–5 TA per group. d The number of well-positioned triads relative to sarcomere number, determined from TEM pictures in TA from wild type and Mtm1−/y mice, showed no significant change from D42 to D84. Black triangles illustrate increasing age (42–84 and 210 days). Mean ± s.e.m. of n = 2–3 TA per group. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; ****P ≤ 0.0001. ##P ≤ 0.01; Mtm1−/y control vs tamoxifen at D42. ns non-significant; Ø no surviving mice. One-way ANOVA followed by Fisher’s LSD post-test

References

    1. Jungbluth H, Gautel M. Pathogenic mechanisms in centronuclear myopathies. Front. Aging Neurosci. 2014;6:339. doi: 10.3389/fnagi.2014.00339.
    1. Lawlor MW, et al. Skeletal muscle pathology in X-Linked myotubular myopathy: review with cross-species comparisons. J. Neuropathol. Exp. Neurol. 2016;75:102–110. doi: 10.1093/jnen/nlv020.
    1. Romero NB. Centronuclear myopathies: a widening concept. Neuromuscul. Disord. 2010;20:223–228. doi: 10.1016/j.nmd.2010.01.014.
    1. Barth PG, Dubowitz V. X-linked myotubular myopathy--a long-term follow-up study. Eur. J. Paediatr. Neurol. 1998;2:49–56. doi: 10.1016/1090-3798(98)01004-9.
    1. Buj-Bello A, Biancalana V, Moutou C, Laporte J, Mandel JL. Identification of novel mutations in the MTM1 gene causing severe and mild forms of X-linked myotubular myopathy. Hum. Mut. 1999;14:320–325. doi: 10.1002/(SICI)1098-1004(199910)14:4<320::AID-HUMU7>;2-O.
    1. Biancalana V, et al. Affected female carriers of MTM1 mutations display a wide spectrum of clinical and pathological involvement: delineating diagnostic clues. Acta Neuropathol. 2017;134:889–904. doi: 10.1007/s00401-017-1748-0.
    1. Blondeau F, et al. Myotubularin, a phosphatase deficient in myotubular myopathy, acts on phosphatidylinositol 3-kinase and phosphatidylinositol 3-phosphate pathway. Hum. Mol. Genet. 2000;9:2223–2229. doi: 10.1093/oxfordjournals.hmg.a018913.
    1. Tronchère H, et al. Production of phosphatidylinositol 5-phosphate by the phosphoinositide 3-phosphatase myotubularin in mammalian cells. J. Biol. Chem. 2004;279:7304–7312. doi: 10.1074/jbc.M311071200.
    1. Taylor GS, Maehama T, Dixon JE. Myotubularin, a protein tyrosine phosphatase mutated in myotubular myopathy, dephosphorylates the lipid second messenger, phosphatidylinositol 3-phosphate. Proc. Natl Acad. Sci. USA. 2000;97:8910–8915. doi: 10.1073/pnas.160255697.
    1. Bachmann C, et al. Cellular, biochemical and molecular changes in muscles from patients with X-linked myotubular myopathy due to MTM1 mutations. Hum. Mol. Genet. 2017;26:320–332.
    1. Elverman M, et al. Long-term effects of systemic gene therapy in a canine model of myotubular myopathy. Muscle Nerve. 2017;56:943–953. doi: 10.1002/mus.25658.
    1. Mack DL, et al. Systemic AAV8-mediated gene therapy drives whole-body correction of myotubular myopathy in dogs. Mol. Ther. 2017;25:839–854. doi: 10.1016/j.ymthe.2017.02.004.
    1. Childers MK, et al. Gene therapy prolongs survival and restores function in murine and canine models of myotubular myopathy. Sci. Transl. Med. 2014;6:220ra210. doi: 10.1126/scitranslmed.3007523.
    1. Buj-Bello A, et al. AAV-mediated intramuscular delivery of myotubularin corrects the myotubular myopathy phenotype in targeted murine muscle and suggests a function in plasma membrane homeostasis. Hum. Mol. Genet. 2008;17:2132–2143. doi: 10.1093/hmg/ddn112.
    1. Cowling BS, et al. Reducing dynamin 2 expression rescues X-linked centronuclear myopathy. J. Clin. Invest. 2014;124:1350–1363. doi: 10.1172/JCI71206.
    1. Tasfaout H, et al. Antisense oligonucleotide-mediated Dnm2 knockdown prevents and reverts myotubular myopathy in mice. Nat. Commun. 2017;8:15661. doi: 10.1038/ncomms15661.
    1. Cowling BS, et al. Amphiphysin (BIN1) negatively regulates dynamin 2 for normal muscle maturation. J. Clin. Invest. 2017;127:4477–4487. doi: 10.1172/JCI90542.
    1. Ismail HM, Dorchies OM, Scapozza L. The potential and benefits of repurposing existing drugs to treat rare muscular dystrophies. Exp. Opin. Orphan Drugs. 2018;6:259–271. doi: 10.1080/21678707.2018.1452733.
    1. Hayes TG. Pharmacologic treatment of male breast cancer. Expert. Opin. Pharmacother. 2009;10:2499–2510. doi: 10.1517/14656560903200634.
    1. Shagufta AhmadI. Tamoxifen a pioneering drug: an update on the therapeutic potential of tamoxifen derivatives. Eur. J. Med. Chem. 2018;143:515–531. doi: 10.1016/j.ejmech.2017.11.056.
    1. Peddi PF. Hormone receptor positive breast cancer: state of the art. Curr. Opin. Obstet. Gynecol. 2018;30:51–54.
    1. Moein MR, Tabibnejad N, Ghasemzadeh J. Beneficial effect of tamoxifen on sperm recovery in infertile men with nonobstructive azoospermia. Andrologia. 2012;44:194–198. doi: 10.1111/j.1439-0272.2011.01163.x.
    1. Chua ME, et al. Revisiting oestrogen antagonists (clomiphene or tamoxifen) as medical empiric therapy for idiopathic male infertility: a meta-analysis. Andrology. 2013;1:749–757. doi: 10.1111/j.2047-2927.2013.00107.x.
    1. Patel DP, Chandrapal JC, Hotaling JM. Hormone-based treatments in subfertile males. Curr. Urol. Rep. 2016;17:56. doi: 10.1007/s11934-016-0612-4.
    1. Kunath F, Keck B, Antes G, Wullich B, Meerpohl J. Tamoxifen for the management of breast events induced by non-steroidal antiandrogens in patients with prostate cancer: a systematic review. BMC Med. 2012;10:96. doi: 10.1186/1741-7015-10-96.
    1. Payne WG, et al. Down-regulating causes of fibrosis with tamoxifen: a possible cellular/molecular approach to treat rhinophyma. Ann. Plast. Surg. 2006;56:301–305. doi: 10.1097/01.sap.0000199155.73000.2f.
    1. van Bommel EF, Hendriksz TR, Huiskes AW, Zeegers AG. Brief communication: tamoxifen therapy for nonmalignant retroperitoneal fibrosis. Ann. Intern. Med. 2006;144:101–106. doi: 10.7326/0003-4819-144-2-200601170-00007.
    1. Walter AW, et al. Tamoxifen and carboplatin for children with low-grade gliomas: a pilot study at St. Jude Children’s Research Hospital. J. Pediatr. Hematol. Oncol. 2000;22:247–251. doi: 10.1097/00043426-200005000-00010.
    1. Lawrence SE, Arnold Faught K, Vethamuthu J, Lawson ML. Beneficial effects of raloxifene and tamoxifen in the treatment of pubertal gynecomastia. J. Pediatr. 2004;145:71–76. doi: 10.1016/j.jpeds.2004.03.057.
    1. Skapek SX, et al. Safety and efficacy of high-dose tamoxifen and sulindac for desmoid tumor in children: results of a Children’s Oncology Group (COG) Phase II Study. Pediatr. Blood. Cancer. 2013;60:1108–1112. doi: 10.1002/pbc.24457.
    1. Kreher NC, Eugster EA, Shankar RR. The use of tamoxifen to improve height potential in short pubertal boys. Pediatrics. 2005;116:1513–1515. doi: 10.1542/peds.2005-0577.
    1. Lapid O., van Wingerden J. J., & Perlemuter L. Tamoxifen therapy for the management of pubertal gynecomastia: a systematic review. J. Pediatr. Endocrinol. Metab. 26, 803–807 (2013).
    1. Derman O, Kanbur N, Kilic I, Kutluk T. Long-term follow-up of tamoxifen treatment in adolescents with gynecomastia. J. Pediatr. Endocrinol. Metab. 2008;21:449–454. doi: 10.1515/JPEM.2008.21.5.449.
    1. Eugster EA, et al. Tamoxifen treatment for precocious puberty in McCune-Albright syndrome: a multicenter trial. J. Pediatr. 2003;143:60–66. doi: 10.1016/S0022-3476(03)00128-8.
    1. de G, et al. Tamoxifen improves final height prediction in girls with McCune-Albright syndrome: a long follow-up. Horm. Res. Paediatr. 2015;84:184–189. doi: 10.1159/000435881.
    1. Dorchies OM, et al. The anticancer drug tamoxifen counteracts the pathology in a mouse model of duchenne muscular dystrophy. Am. J. Pathol. 2013;182:485–504. doi: 10.1016/j.ajpath.2012.10.018.
    1. Gayi E, et al. Repurposing the selective oestrogen receptor modulator tamoxifen for the treatment of Duchenne muscular dystrophy. Chimia. 2018;72:238–240. doi: 10.2533/chimia.2018.238.
    1. Buj-Bello A, et al. The lipid phosphatase myotubularin is essential for skeletal muscle maintenance but not for myogenesis in mice. Proc. Natl Acad. Sci. USA. 2002;99:15060–15065. doi: 10.1073/pnas.212498399.
    1. Al-Qusairi L, et al. Lack of myotubularin (MTM1) leads to muscle hypotrophy through unbalanced regulation of the autophagy and ubiquitin-proteasome pathways. Faseb J. 2013;27:3384–3394. doi: 10.1096/fj.12-220947.
    1. Sabha N, et al. PIK3C2B inhibition improves function and prolongs survival in myotubular myopathy animal models. J. Clin. Invest. 2016;126:3613–3625. doi: 10.1172/JCI86841.
    1. Bevilacqua JA, et al. “Necklace” fibers, a new histological marker of late-onset MTM1-related centronuclear myopathy. Acta Neuropathol. 2008;117:283. doi: 10.1007/s00401-008-0472-1.
    1. Cowling BS, Toussaint A, Muller J, Laporte J. Defective membrane remodeling in neuromuscular diseases: Insights from animal models. PLoS Genet. 2012;8:e1002595. doi: 10.1371/journal.pgen.1002595.
    1. Laporte J, Kress W, Mandel JL. Diagnosis of X-linked myotubular myopathy by detection of myotubularin. Ann. Neurol. 2001;50:42–46. doi: 10.1002/ana.1033.
    1. Dorchies OM, et al. Normal innervation and differentiation of X-linked myotubular myopathy muscle cells in a nerve-muscle coculture system. Neuromuscul. Disord. 2001;11:736–746. doi: 10.1016/S0960-8966(01)00221-8.
    1. Hnia K, et al. Myotubularin controls desmin intermediate filament architecture and mitochondrial dynamics in human and mouse skeletal muscle. J. Clin. Invest. 2011;121:70–85. doi: 10.1172/JCI44021.
    1. Kerr JP, Ward CW, Bloch RJ. Dysferlin at transverse tubules regulates Ca(2+) homeostasis in skeletal muscle. Front. Physiol. 2014;5:89. doi: 10.3389/fphys.2014.00089.
    1. Hofhuis J, et al. Dysferlin mediates membrane tubulation and links T-tubule biogenesis to muscular dystrophy. J. Cell. Sci. 2017;130:841. doi: 10.1242/jcs.198861.
    1. Hnia K, Vaccari I, Bolino A, Laporte J. Myotubularin phosphoinositide phosphatases: cellular functions and disease pathophysiology. Trends Mol. Med. 2012;18:317–327. doi: 10.1016/j.molmed.2012.04.004.
    1. Lawlor MW, et al. Inhibition of activin receptor type IIB increases strength and lifespan in myotubularin-deficient mice. Am. J. Pathol. 2011;178:784–793. doi: 10.1016/j.ajpath.2010.10.035.
    1. Dowling JJ, et al. Myotubular myopathy and the neuromuscular junction: a novel therapeutic approach from mouse models. Dis. Model Mech. 2012;5:852–859. doi: 10.1242/dmm.009746.
    1. Robb SA, et al. Impaired neuromuscular transmission and response to acetylcholinesterase inhibitors in centronuclear myopathies. Neuromuscul. Disord. 2011;21:379–386. doi: 10.1016/j.nmd.2011.02.012.
    1. Al-Qusairi L, et al. T-tubule disorganization and defective excitation-contraction coupling in muscle fibers lacking myotubularin lipid phosphatase. Proc. Natl Acad. Sci. USA. 2009;106:18763–18768. doi: 10.1073/pnas.0900705106.
    1. Al-Qusairi L, Laporte J. T-tubule biogenesis and triad formation in skeletal muscle and implication in human diseases. Skelet. Muscle. 2011;1:26–26. doi: 10.1186/2044-5040-1-26.
    1. Royer B, et al. The myotubularin–amphiphysin 2 complex in membrane tubulation and centronuclear myopathies. EMBO Rep. 2013;14:907–915. doi: 10.1038/embor.2013.119.
    1. Kutchukian C, et al. Phosphatidylinositol 3-kinase inhibition restores Ca2+ release defects and prolongs survival in myotubularin-deficient mice. Proc. Natl. Acad. Sci. USA. 2016;113:14432–14437. doi: 10.1073/pnas.1604099113.
    1. Vogel VG, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes. JAMA. 2006;295:2727–2741. doi: 10.1001/jama.295.23.joc60074.
    1. Van der Weijden-Van Doornik EM, Slot DE, Burtin C, van der Weijden GA. Grip strength in women being treated for breast cancer and receiving adjuvant endocrine therapy: systematic review. Phys. Ther. 2017;97:904–914. doi: 10.1093/ptj/pzx069.
    1. Baltgalvis KA, Greising SM, Warren GL, Lowe DA. Estrogen regulates estrogen receptors and antioxidant gene expression in mouse skeletal muscle. PLoS ONE. 2010;5:e10164. doi: 10.1371/journal.pone.0010164.
    1. Saceda M, et al. Regulation of the estrogen receptor in MCF-7 cells by estradiol. Mol. Endocrinol. 1988;2:1157–1162. doi: 10.1210/mend-2-12-1157.
    1. Lonard DM, Nawaz Z, Smith CL, O’Malley BW. The 26S proteasome is required for estrogen receptor-a and coactivator turnover and for efficient estrogen receptor-a transactivation. Mol. Cell. 2000;5:939–948. doi: 10.1016/S1097-2765(00)80259-2.
    1. Fetalvero KM, et al. Defective autophagy and mTORC1 signaling in myotubularin null mice. Mol. Cell. Biol. 2013;33:98–110. doi: 10.1128/MCB.01075-12.
    1. Totta P, Busonero C, Leone S, Marino M, Acconcia F. Dynamin II is required for 17β-estradiol signaling and autophagy-based ERα degradation. Sci. Rep. 2016;6:23727. doi: 10.1038/srep23727.
    1. Maani, N. et al. Tamoxifen therapy in a murine model of myotubular myopathy. Nat Commun. (2018).
    1. Sarnat HB. Myotubular myopathy: arrest of morphogenesis of myofibres associated with persistence of fetal vimentin and desmin. Four cases compared with fetal and neonatal muscle. Can. J. Neurol. Sci. 1990;17:109–123. doi: 10.1017/S0317167100030304.
    1. Demonbreun AR, McNally EM. Dynamin 2 the rescue for centronuclear myopathy. J. Clin. Invest. 2014;124:976–978. doi: 10.1172/JCI74434.
    1. Pemmaraju N, Munsell MF, Hortobagyi GN, Giordano SH. Retrospective review of male breast cancer patients: analysis of tamoxifen-related side-effects. Ann. Oncol. 2011;23:1471–1474. doi: 10.1093/annonc/mdr459.
    1. Karatas F, et al. Leg cramps associated with tamoxifen use—possible mechanism and treatment recommendations. J. Buon. 2016;21:520.
    1. Lintermans A, et al. A prospective assessment of musculoskeletal toxicity and loss of grip strength in breast cancer patients receiving adjuvant aromatase inhibitors and tamoxifen, and relation with BMI. Breast Cancer Res. Treat. 2014;146:109–116. doi: 10.1007/s10549-014-2986-7.
    1. Bohm J, et al. Altered splicing of the BIN1 muscle-specific exon in humans and dogs with highly progressive centronuclear myopathy. PLoS Genet. 2013;9:e1003430. doi: 10.1371/journal.pgen.1003430.
    1. Dorchies OM, et al. Green tea extract and its major polyphenol (−)-epigallocatechin gallate improve muscle function in a mouse model for Duchenne muscular dystrophy. Am. J. Physiol. Cell. Physiol. 2006;290:C616–C625. doi: 10.1152/ajpcell.00425.2005.
    1. Hibaoui Y, Reutenauer-Patte J, Patthey-Vuadens O, Ruegg UT, Dorchies OM. Melatonin improves muscle function of the dystrophic mdx5Cv mouse, a model for Duchenne muscular dystrophy. J. Pineal Res. 2011;51:163–171. doi: 10.1111/j.1600-079X.2011.00871.x.
    1. Reutenauer J, Dorchies OM, Patthey-Vuadens O, Vuagniaux G, Ruegg UT. Investigation of Debio 025, a cyclophilin inhibitor, in the dystrophic mdx mouse, a model for Duchenne muscular dystrophy. Br. J. Pharmacol. 2008;155:574–584. doi: 10.1038/bjp.2008.285.
    1. Reutenauer-Patte J, Boittin FX, Patthey-Vuadens O, Ruegg UT, Dorchies OM. Urocortins improve dystrophic skeletal muscle structure and function through both PKA- and Epac-dependent pathways. Am. J. Pathol. 2012;180:749–762. doi: 10.1016/j.ajpath.2011.10.038.
    1. De Andrade PBM, et al. Caloric restriction induces energy-sparing alterations in skeletal muscle contraction, fiber composition and local thyroid hormone metabolism that persist during catch-up fat upon refeeding. Front. Physiol. 2015;6:254.
    1. Cowling BS, et al. Increased expression of wild-type or a centronuclear myopathy mutant of dynamin 2 in skeletal muscle of adult mice leads to structural defects and muscle weakness. Am. J. Pathol. 2011;178:2224–2235. doi: 10.1016/j.ajpath.2011.01.054.
    1. Amoasii L, et al. Phosphatase-dead myotubularin ameliorates X-linked centronuclear myopathy phenotypes in mice. PLoS Genet. 2012;8:e1002965. doi: 10.1371/journal.pgen.1002965.
    1. Boittin FX, et al. Ca2+-independent phospholipase A2 enhances store-operated Ca2+ entry in dystrophic skeletal muscle fibers. J. Cell. Sci. 2006;119:3733–3742. doi: 10.1242/jcs.03184.

Source: PubMed

3
Abonnieren