Autophagy activation in COL6 myopathic patients by a low-protein-diet pilot trial

Silvia Castagnaro, Camilla Pellegrini, Massimo Pellegrini, Martina Chrisam, Patrizia Sabatelli, Silvia Toni, Paolo Grumati, Claudio Ripamonti, Loredana Pratelli, Nadir M Maraldi, Daniela Cocchi, Valeria Righi, Cesare Faldini, Marco Sandri, Paolo Bonaldo, Luciano Merlini, Silvia Castagnaro, Camilla Pellegrini, Massimo Pellegrini, Martina Chrisam, Patrizia Sabatelli, Silvia Toni, Paolo Grumati, Claudio Ripamonti, Loredana Pratelli, Nadir M Maraldi, Daniela Cocchi, Valeria Righi, Cesare Faldini, Marco Sandri, Paolo Bonaldo, Luciano Merlini

Abstract

A pilot clinical trial based on nutritional modulation was designed to assess the efficacy of a one-year low-protein diet in activating autophagy in skeletal muscle of patients affected by COL6/collagen VI-related myopathies. Ullrich congenital muscular dystrophy and Bethlem myopathy are rare inherited muscle disorders caused by mutations of COL6 genes and for which no cure is yet available. Studies in col6 null mice revealed that myofiber degeneration involves autophagy defects and that forced activation of autophagy results in the amelioration of muscle pathology. Seven adult patients affected by COL6 myopathies underwent a controlled low-protein diet for 12 mo and we evaluated the presence of autophagosomes and the mRNA and protein levels for BECN1/Beclin 1 and MAP1LC3B/LC3B in muscle biopsies and blood leukocytes. Safety measures were assessed, including muscle strength, motor and respiratory function, and metabolic parameters. After one y of low-protein diet, autophagic markers were increased in skeletal muscle and blood leukocytes of patients. The treatment was safe as shown by preservation of lean:fat percentage of body composition, muscle strength and function. Moreover, the decreased incidence of myofiber apoptosis indicated benefits in muscle homeostasis, and the metabolic changes pointed at improved mitochondrial function. These data provide evidence that a low-protein diet is able to activate autophagy and is safe and tolerable in patients with COL6 myopathies, pointing at autophagy activation as a potential target for therapeutic applications. In addition, our findings indicate that blood leukocytes are a promising noninvasive tool for monitoring autophagy activation in patients.

Trial registration: ClinicalTrials.gov NCT01438788.

Keywords: Bethlem myopathy; Ullrich congenital muscular dystrophy; autophagy; clinical trial; collagen VI; low-protein diet; muscular dystrophies.

Figures

Figure 1.
Figure 1.
Analysis of autophagy in muscle biopsies of BM and UCMD patients before and after LPD trial. Muscle biopsies from 7 adult patients (P1-P7) were taken before (T0) and after 12 mo LPD (T12). (A) Western blot of MAP1LC3B (LC3B) and BECN1 protein levels in lysates of muscle biopsies at T0 and T12. GAPDH was used as a loading control. Data are representative of at least 3 technical replicates. (B) Densitometric quantification of western blots for LC3B and BECN1. LC3B-II and BECN1 protein levels were calculated as relative intensity with respect to GAPDH. Data represent the mean values (3 experiments) for each patient at T0 and T12. P = 0.018 and P = 0.028, respectively, for the comparison of LC3B-II and BECN1 mean values between T0 and T12 (Wilcoxon signed rank test for paired data). (C) Quantitative RT-PCR analysis of MAP1LC3B and BECN1 mRNA levels at T0 and T12. Data represent the mean value (2 experiments) for each patient at T0 and T12. P = 0.018 and P = 0.028, respectively, for the comparison of MAP1LC3B and BECN1 mean fold induction between T0 and T12 (Wilcoxon signed rank test for paired data). (D) Transmission electron micrographs of muscle biopsies after one-y LPD. Representative images of autophagosomes detected in muscle fibers of patient 1 (left panel) and patient 3 (right panel) containing degraded material (left panel, white arrowhead) and a degenerating mitochondrion (right panel). Scale bars: 200 nm. (E) Quantification of myofibers with autophagosomes/autolysosomes, as determined by transmission electron microscopy analysis of muscle biopsies from patients P1-P7 at T0 and T12. The graphs show the percentage of myofibers containing autophagosomes or autolysosomes from 5 different levels for each biopsy. P = 0.016, for the comparison between T0 and T12, following Wilcoxon signed rank test for paired data.
Figure 2.
Figure 2.
TUNEL assay on muscle biopsies of BM and UCMD patients before and after LPD trial. Muscle biopsies from the 7 patients (P1-P7) were analyzed for apoptosis incidence before (T0) and after a 12-mo LPD (T12). (A) Representative micrographs of TUNEL assay on muscle biopsies of patient 2 at T0 (upper panels) and T12 (lower panels). Nuclei were stained in blue with Hoechst, TUNEL-positive nuclei are in red. Scale bar: 50 µm. (B) Quantification of TUNEL-positive nuclei per area in muscle biopsies from patients P1-P7 at T0 and T12. Mean values from 2 technical replicates of TUNEL assay experiments per condition are shown. P = 0.016, following Wilcoxon signed rank test for paired data, for the comparison between mean values at T0 and T12.
Figure 3.
Figure 3.
Clinical parameters of the patients before and after LPD trial. (A-C) Motor function parameters in each of the 6 patients able to walk at T0 and T12. (A) Distance walked in 6 min (6MWD), measured in meters (m). (B) Timed 4-stair climb test (4 steps), measured in seconds (s). P = 0.034, following Wilcoxon signed rank test for paired data, for the comparison between mean values at T0 and T12. (C) Timed 10-min walk test (10 m), measured in seconds (s). (D) Forced vital capacity percentage (FVC, %) in each of the 7 patients. The comparison between the average FVC percentage at T0 and T12 shows a trend toward increase close to significance (P = 0.059) at T12, with respect to baseline visit (T0).
Figure 4.
Figure 4.
Metabolic parameters of the patients before and after the LPD trial. (A) Histogram showing the respiratory quotient (RQ) calculated at T0 and T12 for each patient (P1-P7). RQ values of each patient at T0 and T12 are listed in Table S3. P = 0.031 for the comparison between T0 and T12, following Wilcoxon signed ranked test. (B) Representative 1H Carr-Purcell-Meiboom-Gill NMR spectra of serum of patient 4 at T0, T6 and T12, showing the methyl resonance of alanine (Ala), lactate (Lac) and acetate (Ac). Lac:Ala ratios in sera of all patients at T0, T6 and T12 are reported in Table 3.
Figure 5.
Figure 5.
Autophagy activation after LPD is similar in muscles and in leukocytes of wild-type mice and col6a1 null mice. (A, B) Western blotting of MAP1LC3B/LC3B in protein lysates of tibialis anterior muscles and blood leukocytes (Leuko) from wild-type and col6a1 null (col6a1−/−) mice fed either with standard diet (SD) or low-protein diet (LPD) for 30 d. Each lane corresponds to one animal. For each condition, animals were injected with 0.4 mg/kg/d of colchicine at the end of the diet, to visualize the autophagic flux. RPS6 was used as a loading control. (C, D) Scatter plots showing linear association (dotted line) between LC3B-II relative intensity in tibialis anterior (TA) and leukocytes from western blot analysis as shown in (A and B). Relative protein levels of LC3B-II in muscles and leukocytes were quantified vs RPS6. Each point corresponds to one animal. Pearson coefficients (R) and P values are shown.
Figure 6.
Figure 6.
Analysis of autophagy in blood leukocytes of BM and UCMD patients before and after the LPD trial. (A) Western blot of MAP1LC3B/LC3B and BECN1 levels in protein lysates of blood leukocytes at T0, T6 and T12. For patient 1, blood sample at T6 was not available. ACTB was used as a loading control. Arrowheads point at the specific band of BECN1. Data are representative of 2 technical replicates. (B) Densitometric quantification of western blot for LC3B and BECN1 in leukocyte fractions at T0 and T12. LC3B-II and BECN1 protein levels were calculated as relative intensity with respect to ACTB. Data represent mean value at T0 and T12 for each patient (2 experiments). P = 0.042 and P = 0.062, respectively for the comparison of LC3B-II and BECN1 values between T0 and T12 (Wilcoxon signed rank test for paired data).

References

    1. Bönnemann CG. The collagen VI-related myopathies Ullrich congenital muscular dystrophy and Bethlem myopathy. Handb Clin Neurol 2011; 101:81-96; PMID:21496625;
    1. Scacheri PC, Gillanders EM, Subramony SH, Vedanarayanan V, Crowe CA, Thakore N, Bingler M, Hoffman EP. Novel mutations in collagen VI genes: expansion of the Bethlem myopathy phenotype. Neurology 2002; 58(4):593-602; PMID:11865138;
    1. Merlini L, Martoni E, Grumati P, Sabatelli P, Squarzoni S, Urciuolo A, Ferlini A, Gualandi F, Bonaldo P. Autosomal recessive myosclerosis myopathy is a collagen VI disorder. Neurology 2008; 71(16):1245-53; PMID:18852439;
    1. Bethlem J, Wijngaarden GK. Benign myopathy, with autosomal dominant inheritance. A report on three pedigrees. Brain 1976; 99(1):91-100; PMID:963533;
    1. Jöbsis GJ, Boers JM, Barth PG, de Visser M. Bethlem myopathy: a slowly progressive congenital muscular dystrophy with contractures. Brain 1999; 122(4):649-55; PMID:10219778;
    1. Merlini L, Morandi L, Granata C, Ballestrazzi A. Bethlem myopathy: early-onset benign autosomal dominant myopathy with contractures. Description of two new families. Neuromuscul Disord 1994; 4(5–6):503-11; PMID:7881296
    1. van der Kooi AJ, de Voogt WG, Bertini E, Merlini L, Talim FB, Ben Yaou R, Urtziberea A, de Visser M. Cardiac and pulmonary investigations in Bethlem myopathy. Arch Neurol 2006; 63(11):1617-21; PMID:17101832;
    1. Camacho Vanegas O, Bertini E, Zhang RZ, Petrini S, Minosse C, Sabatelli P, Giusti B, Chu ML, Pepe G. Ullrich scleroatonic muscular dystrophy is caused by recessive mutations in collagen type VI. Proc Natl Acad Sci USA 2001; 98(13):7516-21; PMID:11381124;
    1. Yonekawa T, Nishino I. Ullrich congenital muscular dystrophy: clinicopathological features, natural history and pathomechanism(s). J Neurol Neurosurg Psychiatry 2015; 86(3):280-7; PMID:24938411;
    1. Irwin WA, Bergamin N, Sabatelli P, Reggiani C, Megighian A, Merlini L, Braghetta P, Columbaro M, Volpin D, Bressan GM, et al.. Mitochondrial dysfunction and apoptosis in myopathic mice with collagen VI deficiency. Nat Genet 2003; 35(4):367-71; PMID:14625552;
    1. Angelin A, Tiepolo T, Sabatelli P, Grumati P, Bergamin N, Golfieri C, Mattioli E, Gualandi F, Ferlini A, Merlini L, et al.. Mitochondrial dysfunction in the pathogenesis of Ullrich congenital muscular dystrophy and prospective therapy with cyclosporins. Proc Natl Acad Sci USA 2007; 104(3):991-6; PMID:17215366;
    1. Sabatelli P, Palma E, Angelin A, Squarzoni S, Urciuolo A, Pellegrini C, Tiepolo T, Bonaldo P, Gualandi F, Merlini L, et al.. Critical evaluation of the use of cell cultures for inclusion in clinical trials of patients affected by collagen VI myopathies. J Cell Physiol 2012; 227(7):2927-35; PMID:21953374;
    1. Merlini L, Angelin A, Tiepolo T, Braghetta P, Sabatelli P, Zamparelli A, Ferlini A, Maraldi NM, Bonaldo P, Bernardi P. Cyclosporin A corrects mitochondrial dysfunction and muscle apoptosis in patients with collagen VI myopathies. Proc Natl Acad Sci USA 2008; 105(13):5225-9; PMID:18362356;
    1. Merlini L, Sabatelli P, Armaroli A, Gnudi S, Angelin A, Grumati P, Michelini ME, Franchella A, Gualandi F, Bertini E, et al.. Cyclosporine A in Ullrich congenital muscular dystrophy: long-term results. Oxid Med Cell Longev 2011; 2011:139194; PMID:22028947;
    1. Grumati P, Coletto L, Sabatelli P, Cescon M, Angelin A, Bertaggia E, Blaauw B, Urciuolo A, Tiepolo T, Merlini L, et al.. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat Med 2010; 16(11):1313-20; PMID:21037586;
    1. Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature 2008; 451(7182):1069-75; PMID:18305538;
    1. Schneider JL, Cuervo AM. Autophagy and human disease: emerging themes. Curr Opin Genet Dev 2014; 26:16-23; PMID:24907664;
    1. Merlini L, Nishino I. Consortium for Autophagy in Muscular Dystrophies. 201st ENMC International Workshop: Autophagy in muscular dystrophies–translational approach, 1-3 November 2013, Bussum, The Netherlands. Neuromuscul Disord 2014; 24(6):546-61; PMID:24746377;
    1. Grumati P, Coletto L, Sandri M, Bonaldo P. Autophagy induction rescues muscular dystrophy. Autophagy 2011; 7(4):426-8; PMID:21543891;
    1. Chrisam M, Pirozzi M, Castagnaro S, Blaauw B, Polishchuck R, Cecconi F, Grumati P, Bonaldo P. Reactivation of autophagy by spermidine ameliorates the myopathic defects of collagen VI-null mice. Autophagy 2015; 11(12):2142-52; PMID:26565691;
    1. Bartlett K, Eaton S. Mitochondrial beta-oxidation. Eur J Biochem 2004; 271(3):462-9; PMID:14728673;
    1. Ju JS, Varadhachary AS, Miller SE, Weihl CC. Quantitation of “autophagic flux” in mature skeletal muscle. Autophagy 2010; 6:929-35; PMID:20657169;
    1. Sandri M, Coletto L, Grumati P, Bonaldo P. Misregulation of autophagy and protein degradation systems in myopathies and muscular dystrophies. J Cell Sci 2013; 126(23):5325-33; PMID:24293330;
    1. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med 2013; 368(7):651-62; PMID:23406030;
    1. Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, Metzger D, Reggiani C, Schiaffino S, Sandri M. Autophagy is required to maintain muscle mass. Cell Metab 2009; 10(6):507-15; PMID:19945408;
    1. Carnio S, LoVerso F, Baraibar MA, Longa E, Khan MM, Maffei M, Reischl M, Canepari M, Loefler S, Kern H, et al.. Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell Rep 2014; 8(5):1509-21; PMID:25176656;
    1. Yang L, Licastro D, Cava E, Veronese N, Spelta F, Rizza W, Bertozzi B, Villareal DT, Hotamisligil GS, Holloszy JO, Fontana L. Long-term calorie restriction enhances cellular quality-control processes in human skeletal muscle. Cell Rep 2016; 14(3):422-8; PMID:26774472;
    1. Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006; 443(7113):780-6; PMID:17051204;
    1. Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med 2013; 19(8):983-97; PMID:23921753;
    1. Pepe G, Bertini E, Bonaldo P, Bushby K, Giusti B, de Visser M, Guicheney P, Lattanzi G, Merlini L, Muntoni F, et al.. Bethlem myopathy (BETHLEM) and Ullrich scleroatonic muscular dystrophy: 100th ENMC international workshop, 23-24 November 2001, Naarden, The Netherlands. Neuromuscul Disord 2002; 12(10):984-93; PMID:12467756;
    1. Baumgartner RN, Wayne SJ, Waters DL, Janssen I, Gallagher D, Morley JE. Sarcopenic obesity predicts instrumental activities of daily living disability in the elderly. Obes Res 2004; 12(12):1995-2004; PMID:15687401;
    1. Mandayam S, Mitch WE. Dietary protein restriction benefits patients with chronic kidney disease. Nephrology (Carlton) 2006; 11(1):53-7; PMID:16509933;
    1. van der Ploeg RJ, Fidler V, Oosterhuis HJ. Hand-held myometry: reference values. J Neurol Neurosurg Psychiatry 1991; 54(3):244-7; PMID:2030353;
    1. Merlini L, Mazzone ES, Solari A, Morandi L. Reliability of hand-held dynamometry in spinal muscular atrophy. Muscle Nerve 2002; 26(1):64-70; PMID:12115950;
    1. Merlini L, Solari A, Vita G, Bertini E, Minetti C, Mongini T, Mazzoni E, Angelini C, Morandi L. Role of gabapentin in spinal muscular atrophy: results of a multicenter, randomized Italian study. J Child Neurol 2003; 18(8):537-41; PMID:13677579;
    1. Merlini L, Bertini E, Minetti C, Mongini T, Morandi L, Angelini C, Vita G. Motor function-muscle strength relationship in spinal muscular atrophy. Muscle Nerve 2004; 29(4):548-52; PMID:15052620;
    1. Sabatelli P, Castagnaro S, Tagliavini F, Chrisam M, Sardone F, Demay L, Richard P, Santi S, Maraldi NM, Merlini L, et al.. Aggresome-autophagy involvement in a sarcopenic patient with rigid spine syndrome and a p.C150R mutation in FHL1 gene. Front Aging Neurosci 2014; 6:215; PMID:25191266;
    1. Tagliavini F, Pellegrini C, Sardone F, Squarzoni S, Paulsson M, Wagener R, Gualandi F, Trabanelli C, Ferlini A, Merlini L, et al.. Defective collagen VI α6 chain expression in the skeletal muscle of patients with collagen VI-related myopathies. Biochim Biophys Acta 2014; 1842(9):1604-12; PMID:24907562;
    1. Martoni E, Urciuolo A, Sabatelli P, Fabris M, Bovolenta M, Neri M, Grumati P, D'Amico A, Pane M, Mercuri E, et al.. Identification and characterization of novel collagen VI non-canonical splicing mutations causing Ullrich congenital muscular dystrophy. Hum Mutat 2009; 30(5):E662-72; PMID:19309692;
    1. Lampe AK, Dunn DM, von Niederhausern AC, Hamil C, Aoyagi A, Laval SH, Marie SK, Chu ML, Swoboda K, Muntoni F, et al.. Automated genomic sequence analysis of the three collagen VI genes: applications to Ullrich congenital muscular dystrophy and Bethlem myopathy. J Med Genet 2005; 42(2):108-20; PMID:15689448;
    1. Cairella G, Scalfi L, eds. LARN Livelli di Assunzione di Riferimento di Nutrienti ed energia per la popolazione italiana. IV Revisione. Milano, Italy: SICS, 2014.

Source: PubMed

Подписаться